Glucagon is critical for the maintenance of blood glucose, however nutrient regulation of pancreatic α-cells remains poorly understood. Here, we identified a role of leucine, a well-known β-cell fuel, in the α-cell–intrinsic regulation of glucagon release. In islet perifusion assays, physiologic concentrations of leucine strongly inhibited alanine- and arginine-stimulated glucagon secretion from human and mouse islets under hypoglycemic conditions. Mechanistically, leucine dose-dependently reduced α-cell cAMP, independently of Ca2+, ATP/ADP, or fatty acid oxidation. Leucine also reduced α-cell cAMP in islets treated with somatostatin receptor 2 antagonists or diazoxide, compounds that limit paracrine signaling from β/δ-cells. Studies in dispersed mouse islets confirmed an α-cell–intrinsic effect. The inhibitory effect of leucine on cAMP was mimicked by glucose, α-ketoisocaproate, succinate, and the glutamate dehydrogenase activator BCH and blocked by cyanide, indicating a mechanism dependent on mitochondrial metabolism. Glucose dose-dependently reduced the impact of leucine on α-cell cAMP, indicating an overlap in function; however, leucine was still effective at suppressing glucagon secretion in the presence of elevated glucose, amino acids, and the incretin GIP. Taken together, these findings show that leucine plays an intrinsic role in limiting the α-cell secretory tone across the physiologic range of glucose levels, complementing the inhibitory paracrine actions of β/δ-cells.

Article Highlights
  • Despite the critical role of pancreatic islets as nutrient sensors, the mechanisms by which amino acids regulate glucagon release remain incompletely understood.

  • We show that leucine, a well-known β-cell fuel, regulates glucagon secretion via a combination of α-cell–intrinsic and islet paracrine signaling.

  • Leucine intrinsically suppresses α-cell cAMP, independently of α-cell Ca2+, ATP/ADP, or fatty acid oxidation.

  • Our findings demonstrate complementary roles of leucine and glucose in the regulation of α-cell cAMP and glucagon secretion.

Glucagon secretion from pancreatic islet α-cells plays a crucial role in maintaining blood glucose under fasting and hypoglycemic conditions, and supports insulin secretion in the fed state via signaling to neighboring β-cells (1). Glucagon is released through Ca2+-dependent exocytosis that is regulated by the electrical activity of the α-cell and the endoplasmic reticulum, as well as by cAMP, which acts by directly stimulating the exocytotic machinery and recruiting additional glucagon granules into the readily releasable pool (2). Although many studies establish the respective roles of α-cell Ca2+ and cAMP in regulating the timing and magnitude of glucagon secretion, nutrient regulation of α-cell activity remains poorly understood (2,3).

Glucose is an inhibitor of glucagon secretion that regulates α-cell activity through both paracrine and intrinsic mechanisms. At elevated glucose concentrations, β-cell activation suppresses glucagon secretion by releasing several inhibitory factors that include insulin, GABA, Zn2+, urocortin 3, and serotonin/5-HT (4–8). Insulin, despite reducing α-cell cAMP immunofluorescence (4), was later found to reduce glucagon secretion independently of cAMP (9,10), whereas urocortin 3 is coreleased with insulin and suppresses α-cell cAMP by stimulating somatostatin release from δ-cells (4,11,12,13). Antagonism of somatostatin receptor 2 (Sstr2) increases glucagon secretion at both low and high glucose concentrations, establishing the importance of both tonic- and glucose-stimulated paracrine inhibition by δ-cells (4,9,14). However, glucagon secretion is maximally inhibited by glucose at concentrations that do not stimulate insulin release, including in somatostatin knockout mice (15–17), highlighting the importance of α-cell–intrinsic regulation by glucose (18).

Substantial research on α-cell metabolism has focused on the ability of glycolysis to suppress glucagon secretion (17,19–22). Glucose reinforces its own metabolism by switching off mitochondrial β-oxidation via inhibition of CPT1 fatty acid transporters (23,24). However, the high level of lactate dehydrogenase in α-cells shunts glucose-derived pyruvate away from mitochondria (25–27) such that glycolysis is poorly coupled to oxidative phosphorylation (24,26,28). Part of the glucagonostatic effect of glucose may be due to ATP/ADP or lactate-dependent regulation of the KATP channel (15,19,24,25,27), although glucose also suppresses glucagon secretion in the presence of KATP channel blockers (16,17,29). Consistently, glucose intrinsically suppresses α-cell cAMP (9) and glucagon exocytosis (30,31) independently of Ca2+ regulation. Although these studies establish a crucial role for intermediary metabolism in the regulation of glucagon secretion, a majority have been conducted with glucose as the only substrate.

In addition to glucose, α-cells are sensitive to most amino acids (32,33). Glucagon release is potently stimulated by the electrogenic fuels arginine and alanine, which are thought to depolarize the α-cell plasma membrane and stimulate Ca2+ influx (34–36). Glutamine is not a potent secretagogue but is important for α-cell proliferation (32), whereas glutamate and glycine stimulate glucagon secretion through α-cell membrane receptors (37,38). One amino acid that has not received significant attention is leucine, which is primarily considered a β-cell fuel. Leucine is the only amino acid that is sufficient to stimulate insulin secretion at low glucose. As an oxidative and anaplerotic mitochondrial fuel, leucine metabolism generates acetyl-CoA for oxidation in the TCA cycle and allosterically activates the anaplerotic enzyme glutamate dehydrogenase (GDH) (39,40). Although gain-of-function mutations in GDH cause β-cell hypersecretion and protein-induced hypoglycemia (41), it is not clear whether leucine regulates α-cell activity.

Here, we show that physiologic concentrations of leucine suppress glucagon secretion from both mouse and human islets. Leucine lowers α-cell cAMP, independently of α-cell Ca2+, ATP/ADP ratio, or mitochondrial fatty acid oxidation. Although activation of β/δ-cells contributes to its glucagonostatic effect, leucine also acts directly on the α-cell. The suppressive effect of leucine on cAMP is mimicked by several different mitochondrial fuels and blocked by the mitochondrial poison cyanide. Although glucose also suppresses cAMP, the higher potency of leucine allows it to work more effectively under hypoglycemic and euglycemic conditions. Together, our data suggest a central role of mitochondria in the regulation of glucagon secretion through the α-cell–intrinsic suppression of cAMP.

Human Islets

Human islets were obtained from the Alberta Diabetes Institute Islet Core, Prodo Laboratories, and the Integrated Islet Distribution Program. The age, sex, BMI, and HbA1c percentage of each donor are provided in Supplementary Table 1. Islets were cultured in Pim(R) Islet Media (Prodo Laboratories) for a period of 24–48 h after being received. This study used Integrated Islet Distribution Program data from the Organ Procurement and Transplantation Network. The interpretation and reporting of these data are the responsibility of the authors and in no way should be seen as an official policy of or interpretation by the Organ Procurement and Transplantation Network or the U.S. Government.

Mouse Islets

Wild-type C57BL/6J mice (age 12–15 weeks) were obtained from The Jackson Laboratory. GcgCreERT mice (42) (Jax 030346) were crossed with GCaMP6s mice (Jax 028866), a Cre-dependent Ca2+ indicator strain, and CAMPER mice (Jax 032205), a Cre-dependent cAMP indicator strain. As in our previous study (34), the mice were not treated with tamoxifen (additional details are provided in the next section). Islets were isolated and cultured as in the study by Foster et al. (43) and dispersed as in that by Ho et al. (25). All procedures involving animals were approved by the Institutional Animal Care and Use Committee of the William S. Middleton Memorial Veterans Hospital and followed the National Institutes of Health Guide for the Care and Use of Laboratory Animals.

Imaging of α-Cell ATP/ADP, cAMP, and Ca2+

Reagents were obtained from Sigma-Aldrich unless indicated otherwise. For imaging of α-cell Ca2+, islets from GcgCreERT:GCaMP6s mice were cultured in the presence of 100 nmol/L 4-hydroxytamoxifen and imaged 3 days postisolation. However, we found that 4-hydroxytamoxifen was unnecessary for GCaMP6s expression, provided that GcgCreERT was present in the dam (Supplementary Table 2). Leveraging this breeding strategy to include GcgCreERT in both the sire and dam, islets isolated from GcgCreERT:CAMPER mice were not treated with 4-hydroxytamoxifen. Expression of GCaMP6s and CAMPER was not observed in the absence GcgCreERT. For imaging of α-cell cAMP, islets were isolated from GcgCreERT:CAMPER mice and imaged the next day, except in circumstances shown in Figs. 3G and 6G and Supplementary Fig. 4, where islets from GcgCreERT mice were cultured for 2 h with Cre-dependent CMV-FlexOn-CAMPER adenovirus (VectorBuilder), transferred to media containing 100 nmol/L 4-hydroxytamoxifen, and imaged 2 days postisolation. For imaging of α-cell ATP/ADP, islets from GcgCreERT mice were cultured for 2 h with Cre-dependent CMV-FlexOn-Perceval-HR adenovirus (VectorBuilder), transferred to media containing 100 nmol/L 4-hydroxytamoxifen, and imaged 3 days post-isolation. Islets were imaged in an RC-41LP imaging chamber (Warner Instruments) mounted on a Nikon Ti2 microscope equipped with a 10×/0.5 NA SuperFluor objective (Nikon). The imaging solution contained 137 mmol/L NaCl, 5.6 mmol/L KCl, 1.2 mmol/L MgCl2, 0.5 mmol/L NaH2PO4, 4.2 mmol/L NaHCO3, 10 mmol/L HEPES, and 2.6 mmol/L CaCl2 (pH 7.4). Metabolites, mouse GIP (Pheonix Pharmaceuticals), CYN154806 (Tocris Bioscience), and cyanide were added as indicated. The flow rate was maintained at 0.25 mL/min using a feedback-controlled flow cell (Fluigent MCFS-EZ), and the temperature was maintained at 33°C using solution and chamber heaters (Warner). Filters sets (excitation, dichroic, emission) were as follows: Perceval-HR (511/16, 434/21, FF459/526/596-Di01, FF01-542/27; Semrock), CAMPER (434/21, FF459/526/596-Di01, FF02-470/30 and FF01-542/27; Semrock), and GCaMP6s (ET500/20x, ET-Fura2-GFP, ET535/30m; Chroma). Fluorescence emission was collected with a Photometrics Prime 95B or Hamamatsu ORCA-Flash4.0 CMOS camera every 6 s. A single region of interest was used to quantify the average response of each islet using NIS-Elements software (Nikon), with cAMP levels reported as the CAMPER emission ratio R470/542 normalized to the initial condition (R/Ro), ATP/ADP reported as the Perceval-HR excitation ratio R511/434 (R/Ro), and Ca2+ reported as the fluorescence intensity normalized to the initial condition (F/Fo). Single α-cell quantification of Perceval-HR, CAMPER, and GCaMP6s was performed with the Imaris Spots tool (Andor).

Islet Perifusion Assays

For each assay, islets from 12 mice or from a single human donor were pooled and then divided into a 12-chamber perifusion system (BioRep PERI5) with 78–100 islets per chamber. Each independent experiment is presented on a separate graph, and the number of technical replicates (corresponding to the number of chambers) is indicated in the figure legend for each experimental condition. Islets were equilibrated at 2 mmol/L glucose for 48 min at 37°C in KRPH buffer (140 mmol/L NaCl, 4.7 mmol/L KCl, 1.5 mmol/L CaCl2, 1 mmol/L NaH2PO4, 1 mmol/L MgSO4, 2 mmol/L NaHCO3, 5 mmol/L HEPES, and 0.1% BSA [pH 7.4]) with 100 μL Bio-Gel P-4 (cat. no. 1504124; Bio-Rad) before stimulation, as described in the figure legends. An auxiliary peristaltic pump (Golander Pump), which bypasses the PERI5 pump and manifold, was used in circumstances shown in Figs. 11, 1J, 2E and F, and 7AFSupplementary Figs. 1 and 5A and B. The perifusion flow rate was 200 μL/min, except in circumstances shown in Figs. 1I and J, 2E and F, and 7AF and Supplementary Figs. 1 and 5A and B, when the flow rate was reduced to 110 μL/min to resolve basal glucagon release. Insulin and glucagon immunoassays (Promega CS3037A01 and W8022) were used with a TECAN Spark plate reader.

Fatty Acid Oxidation

Fatty acid oxidation was measured as in the study by Armour et al. (24), with the addition of physiologic levels of glutamine and leucine as indicated in the figure legend.

Statistics

Both males and females were analyzed as indicated in Supplementary Table 1; however, sex was not considered a factor in the statistical analysis. All data are presented as mean ± SEM and were analyzed by area under the curve (AUC). For perifusion assays, AUCs were analyzed using an unpaired t test for two conditions, whereas one-way ANOVA was used when comparing more than two conditions. Imaging data were normalized to the control condition (R/Ro or F/Fo). Unpaired t tests were used when comparing the AUCs between control and experimental conditions from two treatment groups. Paired t tests were used when comparing each imaging condition to the preceding (control) condition (note that ANOVA is inappropriate in this case because only two groups can be compared in a controlled fashion).

Data and Resource Availability

The data sets generated and analyzed during the current study are available from the corresponding author on reasonable request.

Leucine Suppresses Glucagon Secretion From Mouse and Human Islets

Islet perifusion was used to simultaneously assess insulin and glucagon secretion from mouse islets stimulated with glucose (2 and 10 mmol/L) and mixed amino acids provided at three times their physiologic concentrations in the portal circulation (LQAR; i.e., L, 1.5 mmol/L leucine; Q, 1.8 mmol/L glutamine; A, mmol/L 6.3 alanine; R, mmol/L 0.6 arginine) (34). At low glucose, LQAR induced a large biphasic rise in glucagon secretion that was further augmented by 10 nmol/L glucose-dependent insulinotropic peptide (GIP) (Fig. 1A), consistent with the glucagonotropic effects of arginine/alanine and stimulation of the α-cell GIP receptor (34). LQAR- and GIP-stimulated glucagon release was dramatically reduced by elevated glucose (Fig. 1A). The LQAR amino acid mixture was sufficient to stimulate insulin secretion at low glucose and amplified insulin secretion at high glucose (Fig. 1B).

Figure 1

Leucine suppresses glucagon secretion from mouse islets. AJ: Perifusion assays were used to simultaneously measure glucagon and insulin release from mouse islets stimulated with 2 or 10 mmol/L glucose (2G and 10G); amino acids provided at three times the physiologic concentrations (L, 1.5 mmol/L leucine; Q, 1.8 mmol/L glutamine; A, 6.3 mmol/L alanine; R, 0.6 mmol/L arginine) except in I and J, where glutamine was provided at 0.6 mmol/L; and 10 nmol/L GIP. The perifusion flow rate was reduced in I and J from 200 to 110 μL/min to resolve basal glucagon secretion. n = 6 chambers from n = 6 mice per condition (AD, I, and J) or n = 3 chambers from n = 3 mice per condition (EH) with 80–100 mouse islets per chamber. Data are shown as mean ± SEM and AUC for each condition compared with control by Student t test (AF, I, and J) or one-way ANOVA with Sidak multiple comparison test (G and H). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. ns, not significant.

Figure 1

Leucine suppresses glucagon secretion from mouse islets. AJ: Perifusion assays were used to simultaneously measure glucagon and insulin release from mouse islets stimulated with 2 or 10 mmol/L glucose (2G and 10G); amino acids provided at three times the physiologic concentrations (L, 1.5 mmol/L leucine; Q, 1.8 mmol/L glutamine; A, 6.3 mmol/L alanine; R, 0.6 mmol/L arginine) except in I and J, where glutamine was provided at 0.6 mmol/L; and 10 nmol/L GIP. The perifusion flow rate was reduced in I and J from 200 to 110 μL/min to resolve basal glucagon secretion. n = 6 chambers from n = 6 mice per condition (AD, I, and J) or n = 3 chambers from n = 3 mice per condition (EH) with 80–100 mouse islets per chamber. Data are shown as mean ± SEM and AUC for each condition compared with control by Student t test (AF, I, and J) or one-way ANOVA with Sidak multiple comparison test (G and H). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. ns, not significant.

Close modal

Similarly to glucose, leucine strongly suppressed glucagon secretion stimulated by the glucagonotropic amino acid mixture QAR, including when GIP was present (Fig. 1C). As expected, leucine alone was sufficient to stimulate insulin secretion, which was further amplified by QAR and GIP (Fig. 1D). Additionally, at 10 mmol/L glucose, leucine suppressed glucagon secretion in the presence of QAR and GIP (Fig. 1E), while potentiating insulin secretion (Fig. 1F). We further tested leucine in the presence of glutamine, because it could be important for maintaining the glutamate pool, which in β-cells feeds the TCA cycle (39) and in α-cells boosts glucagon secretion via autocrine feedback on glutamate receptors (37). Glutamine had no significant effect on glucagon secretion by itself or in the presence of LQAR (Fig. 1G); however, it boosted insulin secretion approximately fourfold in the presence of LQAR (Fig. 1H). Leucine was effective at suppressing glucagon secretion in response to alanine and arginine, regardless of whether glutamine was present (Fig. 1G). Taken together, these findings suggest an important role of leucine in the negative regulation of α-cells at both low and high glucose concentrations.

In the above assays, it was surprising that we did not observe a glucose-dependent reduction in glucagon secretion, an effect reported by many laboratories (e.g., Lai et al. [18]). Because glucagon release is near the limit of detection in the absence of amino acids, we reduced the perifusion flow rate (from 200 to 110 μL/min) to increase the effective concentration of glucagon measured by the immunoassay. At this reduced flow rate, glucagon secretion was reduced by glucose alone (Fig. 1I). A similar glucagonostatic effect was observed in response to leucine (Fig. 1J). Both glucose and leucine increased insulin secretion in these assays (Supplementary Fig. 1). The experiments using this reduced flow rate are listed in Research Design and Methods.

We next used perifusion assays to test the effect of leucine on primary human islets. Leucine stimulated insulin secretion and suppressed QAR-stimulated glucagon secretion in each of the first four preparations tested (Fig. 2A–D). On a technical note, we found that α-cells from human islets were more sensitive than mouse islets to flow changes, which in three of four preparations (Fig. 2A–C but not 2 D) resulted in artifactual increases in glucagon secretion during fraction collector plate changes. This spurious effect was most apparent when the black control traces, which did not receive leucine, exhibited an apparent increase in glucagon release (denoted by arrows in Fig. 2A–C). Using an auxiliary pump to ensure a constant flow rate, we observed that leucine suppressed QAR-stimulated glucagon secretion in two additional human islet preparations without any technical artifacts (Fig. 2E and F). Because all six experiments were controlled, we conclude that leucine suppresses glucagon secretion in human as well as mouse islets.

Figure 2

Leucine regulates glucagon secretion in primary human islets. AF: Perifusion assays were used to simultaneously measure insulin and glucagon secretion from human islets stimulated with glucose and amino acids as in Fig. 1. Each figure panel represents a single donor for a total of n = 6 human donors (n = 6 chambers per condition, with 90–100 human islets per chamber). Arrows in AC indicate a spurious increase in glucagon during the no-leucine control (black traces) because the perifusion pump stopped for fraction collector plate changes; this artifact was not observed in D. Auxiliary perifusion pump was used in E and F to maintain constant flow. Data are shown as mean ± SEM and AUC for each condition compared with control by Student t test. **P < 0.01, ***P < 0.001, ****P < 0.0001.

Figure 2

Leucine regulates glucagon secretion in primary human islets. AF: Perifusion assays were used to simultaneously measure insulin and glucagon secretion from human islets stimulated with glucose and amino acids as in Fig. 1. Each figure panel represents a single donor for a total of n = 6 human donors (n = 6 chambers per condition, with 90–100 human islets per chamber). Arrows in AC indicate a spurious increase in glucagon during the no-leucine control (black traces) because the perifusion pump stopped for fraction collector plate changes; this artifact was not observed in D. Auxiliary perifusion pump was used in E and F to maintain constant flow. Data are shown as mean ± SEM and AUC for each condition compared with control by Student t test. **P < 0.01, ***P < 0.001, ****P < 0.0001.

Close modal

Ca2+ Is an Unlikely Mechanism for the Glucagonostatic Effect of Leucine

To determine whether leucine suppresses glucagon secretion through α-cell Ca2+, mouse islets were isolated from GcgCreERT:GCaMP6s mice. At low glucose, leucine had only a minor effect, whereas QAR strongly increased α-cell Ca2+ (Fig. 3A). Separating the amino acids, glutamine reduced Ca2+, whereas alanine and arginine strongly increased Ca2+ (Supplementary Fig. 2). These findings are consistent with prior reports stating that arginine and alanine depolarize the α-cell plasma membrane (34–36). Although the effect was small, leucine potentiated the glutamine-stimulated reduction in Ca2+, while increasing the Ca2+ response to alanine and arginine (Supplementary Fig. 2), the condition in which glucagon secretion was maximally suppressed by leucine (Fig. 1). It is therefore unlikely that a Ca2+-dependent mechanism explains the glucagonostatic effect of leucine.

Figure 3

Leucine dose-dependently inhibits α-cell cAMP in mouse islets. AF: Measurements of α-cell Ca2+ (A) or α-cell cAMP (BF) from intact mouse islets stimulated with glucose, amino acids, and GIP as in Fig. 1. Left panels show intact islet averages with individual α-cell responses displayed as a heatmap. Representative single-cell traces are shown in the right panels from three α-cells within a representative islet. Data reflect 273–924 single α-cells and 62–131 islets from n = 3 mice per condition. G: Effect of leucine on α-cell cAMP response to 10 nmol/L GIP. Data reflect 47–53 islets from n = 3 mice per condition. H: Dose response of α-cell cAMP to leucine (0.015–1.5 mmol/L) quantified from islet averages at 2 mmol/L glucose (2G, black), 6 mmol/L glucose (6G, gray), or 10 mmol/L glucose (10G, blue). IC50 at each level of glucose was calculated using best-fit curve. Data reflect 68–73 islets from n = 3 mice per condition. Data are shown as mean ± SEM with AUC for each condition compared with control by Student t test (AG). *P < 0.05, ****P < 0.0001.

Figure 3

Leucine dose-dependently inhibits α-cell cAMP in mouse islets. AF: Measurements of α-cell Ca2+ (A) or α-cell cAMP (BF) from intact mouse islets stimulated with glucose, amino acids, and GIP as in Fig. 1. Left panels show intact islet averages with individual α-cell responses displayed as a heatmap. Representative single-cell traces are shown in the right panels from three α-cells within a representative islet. Data reflect 273–924 single α-cells and 62–131 islets from n = 3 mice per condition. G: Effect of leucine on α-cell cAMP response to 10 nmol/L GIP. Data reflect 47–53 islets from n = 3 mice per condition. H: Dose response of α-cell cAMP to leucine (0.015–1.5 mmol/L) quantified from islet averages at 2 mmol/L glucose (2G, black), 6 mmol/L glucose (6G, gray), or 10 mmol/L glucose (10G, blue). IC50 at each level of glucose was calculated using best-fit curve. Data reflect 68–73 islets from n = 3 mice per condition. Data are shown as mean ± SEM with AUC for each condition compared with control by Student t test (AG). *P < 0.05, ****P < 0.0001.

Close modal

Leucine Dose-Dependently Inhibits α-Cell cAMP

In islets isolated from GcgCreERT:CAMPER mice, α-cell cAMP was strongly suppressed by LQAR when delivered at three times the physiologic concentration (Fig. 3B). LQAR also reduced cAMP at physiologic concentrations (Supplementary Fig. 3A). Single-cell analysis (displayed as a heatmap with one cell per row in Fig. 3B, left) revealed a heterogenous response to LQAR, even within α-cells of the same islet. Pulses of cAMP were observed in a subset of α-cells in the presence of 2 mmol/L glucose alone; these pulses were blocked by leucine (Fig. 3B, right). Mouse GIP, which acts directly on the α-cell via GIP receptors (34), was used as a positive control that raises cAMP (Fig. 3B).

Next, we separated the amino acids to determine their individual versus combined effects on α-cell cAMP. Alanine/arginine caused a transient decrease in α-cell cAMP, whereas leucine/glutamine resulted in a more pronounced cAMP reduction (Fig. 3C and Supplementary Fig. 3B). Leucine alone was sufficient to decrease α-cell cAMP, and the addition of QAR had no further effect (Fig. 3D). The provision of glutamine before leucine did not potentiate the drop in cAMP or change the kinetics of the drop (Fig. 3E and F), consistent with the islet perifusion assays (Fig. 1). Leucine did not affect the magnitude of the α-cell GIP response, suggesting separable mechanisms for cAMP regulation; however, the absolute level of cAMP was reduced in the presence of leucine and GIP (Fig. 3G). These data are consistent with the ability of leucine to reduce glucagon secretion in the presence of QAR and GIP (Fig. 1C).

We next performed dose-response curves to determine the IC50 of leucine at varying levels of glucose. Leucine was applied to intact islets between 15 μmol/L and 1.5 mmol/L, which encompassed the fasting and nonfasting concentrations of leucine as measured from the tail vein of mice (108 and 167 μmol/L, respectively) (18,44), as well as the higher postprandial concentration found in the portal blood (501 μmol/L) (34). As glucose was elevated from 2 to 6 mmol/L, the IC50 of leucine increased from 57 ± 19 μmol/L (n = 68 islets; n = 3 mice) to 440 ± 112 μmol/L (n = 73 islets; n = 3 mice). In the presence of 10 mmol/L glucose, leucine suppressed α-cell cAMP starting at 150 μmol/L, and the IC50 increased to 1,162 ± 872 μmol/L (n = 71 islets; n = 3 mice) (Fig. 3H). Combined, these data emphasize that leucine suppresses α-cell cAMP at physiologic levels and across a range of glucose concentrations.

Leucine and Glucose Regulate α-Cell cAMP and Glucagon Secretion Independently of Sstr2

In mouse islets, prior studies with the Sstr2 antagonist CYN154806 (CYN) demonstrated an important role of somatostatin regulation of the α-cell at both low and high glucose concentrations (9,18). Both effects were confirmed in our experiments with glucose, which was used as a positive control. The addition of 500 nmol/L CYN increased glucagon secretion at low glucose (Fig. 4A). Also in the presence of CYN, glucose elevation stimulated insulin secretion and reduced LQAR-stimulated glucagon secretion (Fig. 4A and B). Therefore, our control data indicate Sstr2-dependent inhibition of glucagon release at low glucose, as well as Sstr2-independent suppression of glucagon secretion at high glucose. Similar to glucose, leucine effectively suppressed alanine- and arginine-stimulated glucagon secretion in the presence of CYN (Fig. 4C, left). A comparable glucagonostatic effect of leucine was observed in the added presence of glutamine (Fig. 4C, right). As in earlier experiments, leucine enabled QAR-stimulated insulin release (Fig. 4D).

Figure 4

Leucine modulates α-cell cAMP via both cell-intrinsic and islet paracrine signaling. AD: Perifusion assays of mouse islets stimulated with glucose, amino acids, and GIP as in Fig. 1, with the inclusion of 500 nmol/L Sstr2 antagonist CYN as indicated. n = 3 chambers from n = 3 mice/condition with 90–100 islets/chamber. EN: Epifluorescence microscopy of α-cell cAMP (EM) or α-cell Ca2+ (N) in intact mouse islets stimulated with glucose, amino acids, and GIP as in Fig. 1, with the inclusion of 500 nmol/L CYN or 200 μmol/L diazoxide (Dz) as indicated. Data reflect 43–94 islets from n = 3 mice per group. O: Representative image of dispersed α-cells expressing CAMPER (left), followed by cAMP time courses (right). Data reflect 141 α-cells from n = 2 mice. Data are shown as mean ± SEM with AUC for each condition compared with control by Student t test (A, B, and EO) or one-way ANOVA with Sidak multiple comparison test (C and D). Although AUCs were calculated for the entire stimulus duration, first-phase glucagon was reduced by glucose in the presence of CYN and LQAR, as indicated by **P < 0.01 in panel A (left). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Figure 4

Leucine modulates α-cell cAMP via both cell-intrinsic and islet paracrine signaling. AD: Perifusion assays of mouse islets stimulated with glucose, amino acids, and GIP as in Fig. 1, with the inclusion of 500 nmol/L Sstr2 antagonist CYN as indicated. n = 3 chambers from n = 3 mice/condition with 90–100 islets/chamber. EN: Epifluorescence microscopy of α-cell cAMP (EM) or α-cell Ca2+ (N) in intact mouse islets stimulated with glucose, amino acids, and GIP as in Fig. 1, with the inclusion of 500 nmol/L CYN or 200 μmol/L diazoxide (Dz) as indicated. Data reflect 43–94 islets from n = 3 mice per group. O: Representative image of dispersed α-cells expressing CAMPER (left), followed by cAMP time courses (right). Data reflect 141 α-cells from n = 2 mice. Data are shown as mean ± SEM with AUC for each condition compared with control by Student t test (A, B, and EO) or one-way ANOVA with Sidak multiple comparison test (C and D). Although AUCs were calculated for the entire stimulus duration, first-phase glucagon was reduced by glucose in the presence of CYN and LQAR, as indicated by **P < 0.01 in panel A (left). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Close modal

In parallel with the glucagon secretion measurements, Sstr2 inhibition strongly increased α-cell cAMP in the presence of 2 mmol/L glucose (Fig. 4E). The further addition of glucose effectively lowered α-cell cAMP in the presence of CYN, consistent with δ-cell independent regulation of α-cell cAMP (9). Similarly, LQAR and leucine decreased α-cell cAMP with CYN present at both low and high glucose (Fig. 4F–H). Collectively, these data suggest both Sstr2-dependent and Sstr2-independent mechanisms for leucine suppression of α-cell cAMP and glucagon secretion.

Leucine Modulates α-Cell cAMP via a Combination of α-Cell Intrinsic and Islet Paracrine Signaling

To test for an α-cell–intrinsic effect of leucine in intact islets, we used the KATP channel opener diazoxide (200 μmol/L) to block Ca2+-dependent exocytosis from α-, β-, and δ-cells. Diazoxide increased α-cell cAMP (Fig. 4I–M) to a similar extent as CYN (Fig. 4E–H), which we interpret as a loss of inhibitory paracrine signaling. Sequential application of CYN and diazoxide was used to evaluate this conclusion. In the presence of 2 mmol/L glucose and CYN, diazoxide further increased cAMP, implying that not all paracrine inhibition of α-cell cAMP occurs via Sstr2 (Supplementary Fig. 4). Additionally, in the presence of 2 mmol/L glucose and diazoxide, CYN further increased cAMP levels, indicating that Sstr2 has some tonic activity at low glucose. These results clarify the impact of CYN and diazoxide on α-cell cAMP and confirm the previously reported role of Sstr2 (18).

LQAR effectively lowered α-cell cAMP in the presence of diazoxide, but not to the same level as in its absence (Fig. 4I), consistent with an α-cell–intrinsic effect of LQAR on cAMP in addition to a paracrine effect. GIP was effective at raising α-cell cAMP in the presence of diazoxide (Fig. 4I), as expected for a direct effect on the α-cell GIP receptor (34). In contrast to leucine, alanine/arginine induced only a transient decrease in cAMP, although this effect was reversed by diazoxide (Fig. 4J) and is therefore likely to be Ca2 + or membrane potential-dependent. Similar effects of alanine/arginine on cAMP were observed at physiologic concentrations (A, 2.1 mmol/L; R, 0.2 mmol/L) (34) (Supplementary Fig. 3B). Leucine, alone or in combination with glutamine, strongly suppressed cAMP in the presence of diazoxide (Fig. 4I–M), suggesting that leucine is effective in the absence of paracrine signaling. This effect is also Ca2+ independent because diazoxide blocked LQAR-stimulated changes in Ca2+ (Fig. 4N). Finally, we used islet dispersion to eliminate paracrine and juxtacrine signaling completely. In isolated α-cells from GcgCreERT:CAMPER mice, leucine inhibited α-cell cAMP, with no further effect of QAR (Fig. 4O). Taken together with the CYN and diazoxide experiments in intact islets, our findings in dispersed α-cells confirm an α-cell–intrinsic effect of leucine that suppresses cAMP.

In Contrast to Glucose, Leucine Has No Effect on α-Cell Fatty Acid Oxidation or ATP/ADP Ratio

Because leucine and glucose inhibit cAMP and glucagon secretion in a similar manner, we next considered whether leucine suppresses fatty acid oxidation and cytosolic ATP/ADP (ATP/ADPc), similarly to glucose (Fig. 5A) (24). To do so, β-oxidation of 3H-labeled palmitate was quantified in isolated mouse islets (23,24). Reproducing the experiment performed in these studies, switching islets from 1 to 5 mmol/L glucose in the presence of nonesterified fatty acids (NEFA; 0.36 mmol/L) reduced fatty acid oxidation (Fig. 5B). However, the further addition of glutamine (0.6 mmol/L), alone or in combination with leucine (0.5 mmol/L), had no effect on fatty acid oxidation at either level of glucose.

Figure 5

Leucine has no effect on α-cell fatty acid oxidation or ATP/ADP ratio. A: Glucose suppression of fatty acid oxidation. B: β-oxidation measurements of 3H-labeled palmitate stimulated with glucose, amino acids (Q, 0.6 mmol/L glutamine; L, 0.5 mmol/L leucine), and NEFA (0.36 mmol/L) as indicated. n = 3–5 mice per group. CF: Epifluorescence microscopy of α-cell cytosolic ATP/ADP in intact mouse islets stimulated with glucose (2G, 2 mmol/L glucose; 10G, 10 mmol/L glucose) and amino acids (L, 0.5 mmol/L leucine; Q, 0.6 mmol/L glutamine). Data reflect 58–63 islets from n = 3 mice per group. Data are shown as mean ± SEM with each group compared by one-way ANOVA with Sidak multiple comparison test (B) or AUC for each condition compared with control by Student t test (CF). *P < 0.05, **P < 0.01, ****P < 0.0001.

Figure 5

Leucine has no effect on α-cell fatty acid oxidation or ATP/ADP ratio. A: Glucose suppression of fatty acid oxidation. B: β-oxidation measurements of 3H-labeled palmitate stimulated with glucose, amino acids (Q, 0.6 mmol/L glutamine; L, 0.5 mmol/L leucine), and NEFA (0.36 mmol/L) as indicated. n = 3–5 mice per group. CF: Epifluorescence microscopy of α-cell cytosolic ATP/ADP in intact mouse islets stimulated with glucose (2G, 2 mmol/L glucose; 10G, 10 mmol/L glucose) and amino acids (L, 0.5 mmol/L leucine; Q, 0.6 mmol/L glutamine). Data reflect 58–63 islets from n = 3 mice per group. Data are shown as mean ± SEM with each group compared by one-way ANOVA with Sidak multiple comparison test (B) or AUC for each condition compared with control by Student t test (CF). *P < 0.05, **P < 0.01, ****P < 0.0001.

Close modal

Next, we tested the effects of leucine and glutamine on ATP/ADPc at both low and high glucose. Islets isolated from GcgCreERT mice and expressing Cre-dependent Perceval-HR were used to investigate ATP/ADPc selectively in α-cells. Although glucose exhibited a biphasic effect, first increasing and then reducing α-cell ATP/ADPc, leucine had no significant effect on ATP/ADPc at 2 or 10 mmol/L glucose (Fig. 5C and D). Glutamine by itself reduced ATP/ADPc to a similar extent as 10 mmol/L glucose, and the effects of glucose and glutamine were additive (Fig. 5E and F). Overall, the lack of correlation between ATP/ADPc and cAMP or glucagon secretion argues against ATP/ADPc as an intermediary signaling mechanism.

Leucine Suppression of α-Cell cAMP Is Mimicked by Mitochondrial Fuels and Blocked by Cyanide

To identify α-cell–intrinsic mechanisms linking leucine to cAMP regulation, we tested a series of mitochondrial fuels in the presence of diazoxide to limit paracrine effects from β/δ-cells (Fig. 6A); 10 mmol/L glucose suppressed α-cell cAMP to a similar extent as 1.5 mmol/L leucine, which had no significant effect when added after glucose (Fig. 6B and C). Application of α-ketoisocaproate (KIC; 10 mmol/L), which is transaminated to leucine (45), had a similar effect on α-cell cAMP as leucine and glucose (Fig. 6D). We next tested methyl-succinate, which undergoes a carbon-neutral exchange for malate before directly stimulating succinate dehydrogenase/complex II. Anaplerosis also occurs when the malate converted to pyruvate by malic enzyme cycles back into the mitochondria through pyruvate carboxylase (Fig. 6A); the low malic enzyme activity in mouse islets weakens this effect (46). Methyl-succinate (10 mmol/L) lowered α-cell cAMP, although to a lesser extent than the other fuels (Fig. 6E). These findings are consistent with a common mitochondrial mode of action. To directly test whether stimulation of mitochondrial anaplerosis is sufficient for cAMP regulation, we applied 2-aminobicyclo(2,2,1)heptane-2-carboxylic acid (BCH; 10 mmol/L), a nonmetabolizable leucine analog that activates GDH (40,47). BCH was sufficient to reduce α-cell cAMP, suggesting that GDH activation rather than acetyl-CoA generation underlies leucine suppression of cAMP (Fig. 6F). Although leucine had no effect when applied after BCH, this may be attributed to the off-target inhibition of the β-cell leucine transporter (47), making it difficult to ascertain whether the entire effect of leucine is via GDH. Taken together, these data suggest that mitochondrial metabolism facilitates the α-cell–intrinsic cAMP regulation by leucine, a mechanism that complements extrinsic α-cell regulation via β/δ-cells. Consistent with this interpretation, poisoning mitochondria with 5 mmol/L potassium cyanide (complex IV inhibitor) blocked the suppression of cAMP by leucine (Fig. 6G).

Figure 6

Leucine suppression of α-cell cAMP is mimicked by mitochondrial fuels and blocked by cyanide. A: Mitochondrial fuel metabolism. Glucose fuels the TCA cycle oxidatively via pyruvate dehydrogenase (PDH) and anaplerotically via pyruvate carboxylase (PC), while leucine oxidatively generates acetyl-CoA and anaplerotically stimulates the TCA cycle via GDH. The nonmetabolized leucine BCH stimulates anaplerosis via GDH. Methyl-succinate enters the TCA cycle via exchange with malate for a net carbon neutral exchange to oxidatively stimulate succinate dehydrogenase (SDH)/complex II (CII), with anaplerosis occurring via PC after malate conversion to pyruvate. BG: Epifluorescence microscopy of α-cell cAMP in intact mouse islets represented as islet averages, with individual α-cell responses displayed as a heatmap. Islets were imaged in 2 mmol/L glucose (2G) and 200 μmol/L Dz, with 1.5 mmol/L leucine (L), 10 mmol/L glucose (10G), 10 mmol/L KIC, 10 mmol/L methyl-succinate (Succ), 10 mmol/L BCH, or 5 mmol/L potassium cyanide (KCN) as indicated. Data reflect 82–177 single α-cells and 39–65 islets from n = 3 mice per condition. Data are shown as mean ± SEM with AUC for each condition compared with control by Student t test. **P < 0.01, ****P < 0.0001.

Figure 6

Leucine suppression of α-cell cAMP is mimicked by mitochondrial fuels and blocked by cyanide. A: Mitochondrial fuel metabolism. Glucose fuels the TCA cycle oxidatively via pyruvate dehydrogenase (PDH) and anaplerotically via pyruvate carboxylase (PC), while leucine oxidatively generates acetyl-CoA and anaplerotically stimulates the TCA cycle via GDH. The nonmetabolized leucine BCH stimulates anaplerosis via GDH. Methyl-succinate enters the TCA cycle via exchange with malate for a net carbon neutral exchange to oxidatively stimulate succinate dehydrogenase (SDH)/complex II (CII), with anaplerosis occurring via PC after malate conversion to pyruvate. BG: Epifluorescence microscopy of α-cell cAMP in intact mouse islets represented as islet averages, with individual α-cell responses displayed as a heatmap. Islets were imaged in 2 mmol/L glucose (2G) and 200 μmol/L Dz, with 1.5 mmol/L leucine (L), 10 mmol/L glucose (10G), 10 mmol/L KIC, 10 mmol/L methyl-succinate (Succ), 10 mmol/L BCH, or 5 mmol/L potassium cyanide (KCN) as indicated. Data reflect 82–177 single α-cells and 39–65 islets from n = 3 mice per condition. Data are shown as mean ± SEM with AUC for each condition compared with control by Student t test. **P < 0.01, ****P < 0.0001.

Close modal

Leucine and BCH Suppress Glucagon Release at Concentrations That Do Not Affect Insulin Secretion

Because our initial hormone secretion experiments were carried out in the presence of 1.5 mmol/L leucine (Figs. 1 and 2), a dose that maximally suppresses cAMP (Fig. 3H), we were motivated to evaluate the islet response at lower concentrations. At low glucose, 0.1 and 0.5 mmol/L leucine dose-dependently inhibited glucagon secretion from mouse islets stimulated with alanine and arginine, respectively (Fig. 7A). Although 0.5 mmol/L leucine stimulated insulin secretion under these conditions, 0.1 mmol/L leucine did not, and neither concentration of leucine stimulated insulin release during the application of alanine and arginine. These effects are consistent with the direct effect of leucine on α-cells (Fig. 4). Similar conclusions were reached using the GDH activator BCH, which dose-dependently suppressed glucagon secretion stimulated by alanine and arginine when applied at 0.5 and 2.5 mmol/L, respectively, without stimulating insulin release (Fig. 7B). At the higher dose of BCH used in Fig. 6 (10 mmol/L), insulin secretion was robustly stimulated, and glucagon was strongly suppressed (Supplementary Fig. 5). Finally, we tested the effect of 0.1 and 0.5 mmol/L leucine on four human islet preparations (Fig. 7C–F), although the responses were not as uniform as the 1.5 mmol/L dose (Fig. 2). In the first preparation, insulin was stimulated by 0.5 mmol/L leucine without any effect on glucagon release (Fig. 7C). In the next two preparations, a glucagonostatic effect was observed only at 0.5 mmol/L leucine, and in these cases, insulin release was also stimulated (Fig. 7D and E). In the final preparation, leucine dose-dependently inhibited glucagon secretion in response to alanine and arginine without affecting insulin secretion (Fig. 7F), matching the results in mouse islets. Overall, these data support the conclusion that physiologically relevant concentrations of leucine suppress glucagon secretion. It is also clear, in mouse islets, that β-cell stimulation is a complementary mechanism to the intrinsic regulation of α-cells by leucine.

Figure 7

Leucine suppresses glucagon secretion from mouse and human islets at physiologic concentrations. AF: Perifusion assays were used to simultaneously measure glucagon and insulin release from mouse islets (A and B) or human islets (CF) stimulated with 2 mmol/L glucose (2G), leucine applied at 0.1 mmol/L (0.1 L) or 0.5 mmol/L (0.5 L), BCH applied at 0.5 mmol/L (0.5BCH) or 2.5 mmol/L (2.5BCH), and amino acids (Q, 1.8 mmol/L glutamine; A, 6.3 mmol/L alanine; R, 0.6 mmol/L arginine) as indicated. Mouse perifusion data reflect n = 4 chambers from n = 4 mice per condition with 90–100 islets/chamber (A and B). Human perifusion data reflect n = 4 human preparations (n = 4 chambers per condition with 78–100 human islets per chamber), with each donor displayed on an independent graph (CF). Data are shown as mean ± SEM, and AUC was analyzed for each condition. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Figure 7

Leucine suppresses glucagon secretion from mouse and human islets at physiologic concentrations. AF: Perifusion assays were used to simultaneously measure glucagon and insulin release from mouse islets (A and B) or human islets (CF) stimulated with 2 mmol/L glucose (2G), leucine applied at 0.1 mmol/L (0.1 L) or 0.5 mmol/L (0.5 L), BCH applied at 0.5 mmol/L (0.5BCH) or 2.5 mmol/L (2.5BCH), and amino acids (Q, 1.8 mmol/L glutamine; A, 6.3 mmol/L alanine; R, 0.6 mmol/L arginine) as indicated. Mouse perifusion data reflect n = 4 chambers from n = 4 mice per condition with 90–100 islets/chamber (A and B). Human perifusion data reflect n = 4 human preparations (n = 4 chambers per condition with 78–100 human islets per chamber), with each donor displayed on an independent graph (CF). Data are shown as mean ± SEM, and AUC was analyzed for each condition. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Close modal

Our findings reveal a role of leucine in the regulation of α-cell cAMP and glucagon secretion. Like glucose (9), leucine exerts a direct suppressive effect on α-cell cAMP, in addition to stimulating inhibitory paracrine signals from β/δ-cells. Although the IC50 of leucine increases with glucose, it remains relevant to the level of leucine in the portal circulation (34). While these ex vivo studies may not precisely match the in vivo situation, they suggest that leucine levels in the blood help to set the tone of glucagon secretion in the fasted state and under hypoglycemic conditions, whereas glucose and leucine share control over glucagon secretion in the fed state. Consistently, leucine suppressed glucagon secretion in the presence of GIP, which directly stimulates the α-cell and boosts β-cell function at elevated glucose (34). Most likely, leucine and GIP act on cAMP via separable mechanisms, dependent on metabolic and GPCR signaling, respectively. As discussed below, multiple lines of evidence suggest that leucine works via a mitochondrial mode of action.

Our data indicate that leucine regulates α-cells via a combination of α-cell–intrinsic and islet paracrine signaling, similarly to glucose (4,9,18). Leucine was found to extrinsically regulate α-cells via Sstr2, as expected for a β-cell fuel, whereas mitochondrial fuels, including leucine, BCH, KIC, methyl-succinate, and glucose, were each sufficient to suppress α-cell cAMP in the presence of diazoxide, indicating an intrinsic effect that was confirmed in isolated α-cells. Because diazoxide lowered α-cell Ca2+, these experiments argue against Ca2+-dependent phosphodiesterases or adenylate cyclases as a central mechanism by which mitochondrial fuels reduce α-cell cAMP. The GDH activator BCH, a purely anaplerotic fuel that is not metabolized to acetyl-CoA, was sufficient to suppress α-cell cAMP in the presence of diazoxide. The fact that glucose, which cannot activate GDH, had the same suppressive effect on cAMP suggests a general role of mitochondria in controlling cAMP production and setting α-cell tone. This interpretation is consistent with the ability of cyanide to block the effect of leucine on cAMP.

Although five different mitochondrial fuels suppressed α-cell cAMP (i.e., leucine, BCH, KIC, methyl-succinate, glucose), the link between fuel metabolism and cAMP remains elusive. From the evolutionary perspective, elevated cAMP is a universal cellular signal for energy deficit. It follows that cAMP levels should be low when fuel is replete. The best-known signaling mechanism for such cAMP regulation is the insulin/glucagon ratio that acts via GPCR signaling. However, as previously reported by the Tengholm laboratory (9), and supported by our studies, there may also be a cell-intrinsic mechanism for fuel surfeit to suppress cAMP that remains undiscovered. Ultimately, the role of mitochondrial metabolism must be clarified by generating α-cell knockouts of critical mitochondrial genes (e.g., Glud1, Pc, Pck2). Pdk4 overexpression in α-cells, used as a method to suppress pyruvate dehydrogenase, blocked glucose suppression of glucagon secretion, but cAMP remains to be examined (24). We note that mitochondria may not be the only mechanism for cAMP regulation by leucine, since mTOR signaling plays an important role in amino acid sensing (3), at least chronically (48).

α-Cells are remarkably sensitive to their environment, highlighting the importance of small differences between studies. It was previously reported that glucose elevation (from 1 to 5 mmol/L) reduced α-cell ATP/ADPc in the presence of NEFA and increases ATP/ADPc in its absence (24). However, the same study showed it is possible to avoid endogenous fatty acid depletion by preincubation with >1 mmol/L glucose; in this case, glucose elevation (from 3 to 20 mmol/L) reduced ATP/ADPc in the absence of exogenous NEFA. Similarly, using 2 mmol/L glucose preincubation, we observed a glucose-dependent reduction in ATP/ADPc, Ca2+ (not shown), and glucagon secretion in the absence of exogenous NEFA. In addition to environmental conditions, there may also be species differences that make it challenging to compare between studies. Prior studies examined the role of leucine in the perfused rat pancreas, with mixed results; a synopsis is provided by Leclercq-Meyer et al. (49). There is general agreement that leucine is inhibitory to glucagon output when β-cells are active; however, in contrast to our study, it was observed in the rat pancreas that 0.2 mmol/L leucine was stimulatory to glucagon output at low glucose. Ultimately, the studies are difficult to compare because rat islets are sensitive to glutamine deprivation, which was withheld by Leclercq-Meyer et al. (49). In mouse islets, leucine suppressed cAMP and glucagon secretion whether or not glutamine was present.

As a technical consideration, we successfully used GcgCreERT mice (42) without using tamoxifen. Although it should be possible to perform tamoxifen-inducible studies if GcgCreERT is present in the sire, spurious GCaMP6s fluorescence was observed in the absence of tamoxifen (or 4-hydroxytamoxifen) when GcgCreERT was present in the dam (Supplementary Table 2). Because tamoxifen itself is not inert (50,51), this feature is potentially advantageous for gene knockout; investigators using GcgCreERT mice should tailor the breeding strategy to the experimental goals.

This article contains supplementary material online at https://doi.org/10.2337/figshare.26018686.

Acknowledgments. The authors thank the organ donors and their families for their kind gifts in support of diabetes research, as well as the Human Organ Procurement and Exchange program and Trillium Gift of Life Network for their work in procuring human donor pancreata for research and Dr. Patrick MacDonald’s team (Alberta Diabetes Institute) for their efforts in human islet isolation. Graphics were created using BioRender.com.

Funding. The Merrins laboratory is supported by the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH; R01DK113103 and R01DK127637), IIDP (BS587P), and U.S. Department of Veterans Affairs Biomedical Laboratory Research and Development Service (I01BX005113) (M.J.M.). E.R.K. received a predoctoral fellowship from the NIH/NIDDK (F31DK134171). The Knudsen laboratory is supported by a Novo Nordisk Foundation Excellence Emerging Investigator Grant in Endocrinology and Metabolism (0054300) (J.G.K.) Human pancreatic islets were provided in part by the NIDDK-funded IIDP (RRID:SCR_014387) at City of Hope (UC4DK098085) and the JDRF-funded IIDP Islet Award Initiative.

This work used facilities and resources from the William S. Middleton Memorial Veterans Hospital and does not represent the views of the Department of Veterans Affairs or the U.S. Government.

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

Author Contributions. E.R.K., H.R.F., E.J., and M.H.E. performed experiments. E.R.K. and M.J.M. conceptualized the studies and drafted the manuscript. J.G.K. contributed resources. All authors reviewed/edited the manuscript. M.J.M. 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. Parts of this study were presented in abstract form at the 66th Biophysical Society Annual Meeting, San Francisco, CA, 19–23 February 2022.

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.
Acreman
S
,
Zhang
Q
.
Regulation of α-cell glucagon secretion: the role of second messengers
.
Chronic Dis Transl Med
2022
;
8
:
7
18
3.
Armour
SL
,
Stanley
JE
,
Cantley
J
,
Dean
ED
,
Knudsen
JG
.
Metabolic regulation of glucagon secretion
.
J Endocrinol
2023
;
259
:
e230081
.
4.
Elliott
AD
,
Ustione
A
,
Piston
DW
.
Somatostatin and insulin mediate glucose-inhibited glucagon secretion in the pancreatic α-cell by lowering cAMP
.
Am J Physiol-Endocrinol Metab
2015
;
308
:
E130
E143
5.
Wendt
A
,
Birnir
B
,
Buschard
K
, et al
.
Glucose inhibition of glucagon secretion from rat α-cells is mediated by GABA released from neighboring β-cells
.
Diabetes
2004
;
53
:
1038
1045
6.
Ishihara
H
,
Maechler
P
,
Gjinovci
A
,
Herrera
P-L
,
Wollheim
CB
.
Islet β-cell secretion determines glucagon release from neighbouring α-cells
.
Nat Cell Biol
2003
;
5
:
330
335
7.
Rorsman
P
,
Berggren
PO
,
Bokvist
K
, et al
.
Glucose-inhibition of glucagon secretion involves activation of GABAA-receptor chloride channels
.
Nature
1989
;
341
:
233
236
8.
Almaça
J
,
Molina
J
,
Menegaz
D
, et al
.
Human beta cells produce and release serotonin to inhibit glucagon secretion from alpha Cells
.
Cell Rep
2016
;
17
:
3281
3291
9.
Yu
Q
,
Shuai
H
,
Ahooghalandari
P
,
Gylfe
E
,
Tengholm
A
.
Glucose controls glucagon secretion by directly modulating cAMP in alpha cells
.
Diabetologia
2019
;
62
:
1212
1224
10.
Vergari
E
,
Knudsen
JG
,
Ramracheya
R
, et al
.
Insulin inhibits glucagon release by SGLT2-induced stimulation of somatostatin secretion
.
Nat Commun
2019
;
10
:
139
11.
Rorsman
P
,
Huising
MO
.
The somatostatin-secreting pancreatic δ-cell in health and disease
.
Nat Rev Endocrinol
2018
;
14
:
404
414
12.
van der Meulen
T
,
Donaldson
CJ
,
Cáceres
E
, et al
.
Urocortin3 mediates somatostatin-dependent negative feedback control of insulin secretion
.
Nat Med
2015
;
21
:
769
776
13.
Noguchi
GM
,
Castillo
VC
,
Donaldson
CJ
, et al
.
Urocortin 3 contributes to paracrine inhibition of islet alpha cells in mice
.
J Endocrinol
2024
;
261
:
e240018
14.
Kailey
B
,
van de Bunt
M
,
Cheley
S
, et al
.
SSTR2 is the functionally dominant somatostatin receptor in human pancreatic β- and α-cells
.
Am J Physiol-Endocrinol Metab
2012
;
303
:
E1107
E1116
15.
MacDonald
PE
,
De Marinis
YZ
,
Ramracheya
R
, et al
.
A KATP channel-dependent pathway within α cells regulates glucagon release from both rodent and human islets of Langerhans
.
PLoS Biol
2007
;
5
:
e143
16.
Vieira
E
,
Salehi
A
,
Gylfe
E
.
Glucose inhibits glucagon secretion by a direct effect on mouse pancreatic alpha cells
.
Diabetologia
2007
;
50
:
370
379
17.
Cheng-Xue
R
,
Gómez-Ruiz
A
,
Antoine
N
, et al
.
Tolbutamide controls glucagon release from mouse islets differently than glucose: involvement of K(ATP) channels from both α-cells and δ-cells
.
Diabetes
2013
;
62
:
1612
1622
18.
Lai
B-K
,
Chae
H
,
Gómez-Ruiz
A
, et al
.
Somatostatin is only partly required for the glucagonostatic effect of glucose but is necessary for the glucagonostatic effect of KATP channel blockers
.
Diabetes
2018
;
67
:
2239
2253
19.
Zhang
Q
,
Ramracheya
R
,
Lahmann
C
, et al
.
Role of KATP channels in glucose-regulated glucagon secretion and impaired counterregulation in type 2 diabetes
.
Cell Metab
2013
;
18
:
871
882
20.
Bahl
V
,
Lee May
C
,
Perez
A
,
Glaser
B
,
Kaestner
KH
.
Genetic activation of α-cell glucokinase in mice causes enhanced glucose-suppression of glucagon secretion during normal and diabetic states
.
Mol Metab
2021
;
49
:
101193
21.
Moede
T
,
Leibiger
B
,
Vaca Sanchez
P
, et al
.
Glucokinase intrinsically regulates glucose sensing and glucagon secretion in pancreatic alpha cells
.
Sci Rep
2020
;
10
:
20145
22.
Basco
D
,
Zhang
Q
,
Salehi
A
, et al
.
α-Cell glucokinase suppresses glucose-regulated glucagon secretion
.
Nat Commun
2018
;
9
:
546
23.
Briant
LJB
,
Dodd
MS
,
Chibalina
MV
, et al
.
CPT1a-dependent long-chain fatty acid oxidation contributes to maintaining glucagon secretion from pancreatic islets
.
Cell Rep
2018
;
23
:
3300
3311
24.
Armour
SL
,
Frueh
A
,
Chibalina
MV
, et al
.
Glucose Controls glucagon secretion by regulating fatty acid oxidation in pancreatic α-cells
.
Diabetes
2023
;
72
:
1446
1459
25.
Ho
T
,
Potapenko
E
,
Davis
DB
,
Merrins
MJ
.
A plasma membrane-associated glycolytic metabolon is functionally coupled to KATP channels in pancreatic α and β cells from humans and mice
.
Cell Rep
2023
;
42
:
112394
26.
Schuit
F
,
De Vos
A
,
Farfari
S
, et al
.
Metabolic fate of glucose in purified islet cells. Glucose-regulated anaplerosis in beta cells
.
J Biol Chem
1997
;
272
:
18572
18579
27.
Zaborska
KE
,
Dadi
PK
,
Dickerson
MT
, et al
.
Lactate activation of α-cell KATP channels inhibits glucagon secretion by hyperpolarizing the membrane potential and reducing Ca2+ entry
.
Mol Metab
2020
;
42
:
101056
28.
Wang
Z
,
Gurlo
T
,
Matveyenko
AV
, et al
.
Live-cell imaging of glucose-induced metabolic coupling of β and α cell metabolism in health and type 2 diabetes
.
Commun Biol
2021
;
4
:
594
29.
Le Marchand
SJ
,
Piston
DW
.
Glucose decouples intracellular Ca2+ activity from glucagon secretion in mouse pancreatic islet alpha-cells
.
PLoS One
2012
;
7
:
e47084
30.
Omar-Hmeadi
M
,
Lund
P-E
,
Gandasi
NR
,
Tengholm
A
,
Barg
S
.
Paracrine control of α-cell glucagon exocytosis is compromised in human type-2 diabetes
.
Nat Commun
2020
;
11
:
1896
31.
Dai
X-Q
,
Camunas-Soler
J
,
Briant
LJB
, et al
.
Heterogenous impairment of α cell function in type 2 diabetes is linked to cell maturation state
.
Cell Metab
2022
;
34
:
256
268.e5
32.
Dean
ED
.
A primary role for α-cells as amino acid sensors
.
Diabetes
2020
;
69
:
542
549
33.
Rocha
DM
,
Faloona
GR
,
Unger
RH
.
Glucagon-stimulating activity of 20 amino acids in dogs
.
J Clin Invest
1972
;
51
:
2346
2351
34.
El
K
,
Gray
SM
,
Capozzi
ME
, et al
.
GIP mediates the incretin effect and glucose tolerance by dual actions on α cells and β cells
.
Sci Adv
2021
;
7
:
eabf1948
.
35.
Gerich
JE
,
Frankel
BJ
,
Fanska
R
,
West
L
,
Forsham
PH
,
Grodsky
GM
.
Calcium dependency of glucagon secretion from the in vitro perfused rat pancreas
.
Endocrinology
1974
;
94
:
1381
1385
36.
Rorsman
P
,
Hellman
B
.
Voltage-activated currents in guinea pig pancreatic alpha 2 cells. Evidence for Ca2+-dependent action potentials
.
J Gen Physiol
1988
;
91
:
223
242
37.
Cabrera
O
,
Jacques-Silva
MC
,
Speier
S
, et al
.
Glutamate is a positive autocrine signal for glucagon release
.
Cell Metab
2008
;
7
:
545
554
38.
Li
C
,
Liu
C
,
Nissim
I
, et al
.
Regulation of glucagon secretion in normal and diabetic human islets by γ-hydroxybutyrate and glycine
.
J Biol Chem
2013
;
288
:
3938
3951
39.
Li
C
,
Najafi
H
,
Daikhin
Y
, et al
.
Regulation of leucine-stimulated insulin secretion and glutamine metabolism in isolated rat islets
.
J Biol Chem
2003
;
278
:
2853
2858
40.
Sener
A
,
Malaisse
WJ
.
L-leucine and a nonmetabolized analogue activate pancreatic islet glutamate dehydrogenase
.
Nature
1980
;
288
:
187
189
41.
Stanley
CA
,
Lieu
YK
,
Hsu
BY
, et al
.
Hyperinsulinism and hyperammonemia in infants with regulatory mutations of the glutamate dehydrogenase gene
.
N Engl J Med
1998
;
338
:
1352
1357
42.
Ackermann
AM
,
Zhang
J
,
Heller
A
,
Briker
A
,
Kaestner
KH
.
High-fidelity glucagon-CreER mouse line generated by CRISPR-Cas9 assisted gene targeting
.
Mol Metab
2017
;
6
:
236
244
43.
Foster
HR
,
Ho
T
,
Potapenko
E
, et al
.
β-cell deletion of the PKm1 and PKm2 isoforms of pyruvate kinase in mice reveals their essential role as nutrient sensors for the KATP channel
.
Elife
2022
;
11
:
e79422
44.
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
45.
Zhou
Y
,
Jetton
TL
,
Goshorn
S
,
Lynch
CJ
,
She
P
.
Transamination is required for {alpha}-ketoisocaproate but not leucine to stimulate insulin secretion
.
J Biol Chem
2010
;
285
:
33718
33726
46.
MacDonald
MJ
.
Differences between mouse and rat pancreatic islets: succinate responsiveness, malic enzyme, and anaplerosis
.
Am J Physiol Endocrinol Metab
2002
;
283
:
E302
E310
47.
Cheng
Q
,
Beltran
VD
,
Chan
SMH
,
Brown
JR
,
Bevington
A
,
Herbert
TP
.
System-L amino acid transporters play a key role in pancreatic β-cell signalling and function
.
J Mol Endocrinol
2016
;
56
:
175
187
48.
Riahi
Y
,
Kogot-Levin
A
,
Kadosh
L
, et al
.
Hyperglucagonaemia in diabetes: altered amino acid metabolism triggers mTORC1 activation, which drives glucagon production
.
Diabetologia
2023
;
66
:
1925
1942
49.
Leclercq-Meyer
V
,
Marchand
J
,
Woussen-Colle
MC
,
Giroix
MH
,
Malaisse
WJ
.
Multiple effects of leucine on glucagon, insulin, and somatostatin secretion from the perfused rat pancreas
.
Endocrinology
1985
;
116
:
1168
1174
50.
Ahn
S-H
,
Granger
A
,
Rankin
MM
,
Lam
CJ
,
Cox
AR
,
Kushner
JA
.
Tamoxifen suppresses pancreatic β-cell proliferation in mice
.
PLoS One
2019
;
14
:
e0214829
51.
Carboneau
BA
,
Le
TDV
,
Dunn
JC
,
Gannon
M
.
Unexpected effects of the MIP-CreER transgene and tamoxifen on β-cell growth in C57Bl6/J male mice
.
Physiol Rep
2016
;
4
:
e12863
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.