Liver-specific disruption of the murine glucagon receptor produces α-cell hyperplasia: evidence for a circulating α-cell growth factor

Christine Longuet, Ana M Robledo, E Danielle Dean, Chunhua Dai, Safina Ali, Ian McGuinness, Vincent de Chavez, Patricia M Vuguin, Maureen J Charron, Alvin C Powers, Daniel J Drucker, Christine Longuet, Ana M Robledo, E Danielle Dean, Chunhua Dai, Safina Ali, Ian McGuinness, Vincent de Chavez, Patricia M Vuguin, Maureen J Charron, Alvin C Powers, Daniel J Drucker

Abstract

Glucagon is a critical regulator of glucose homeostasis; however, mechanisms regulating glucagon action and α-cell function and number are incompletely understood. To elucidate the role of the hepatic glucagon receptor (Gcgr) in glucagon action, we generated mice with hepatocyte-specific deletion of the glucagon receptor. Gcgr(Hep)(-/-) mice exhibited reductions in fasting blood glucose and improvements in insulin sensitivity and glucose tolerance compared with wild-type controls, similar in magnitude to changes observed in Gcgr(-/-) mice. Despite preservation of islet Gcgr signaling, Gcgr(Hep)(-/-) mice developed hyperglucagonemia and α-cell hyperplasia. To investigate mechanisms by which signaling through the Gcgr regulates α-cell mass, wild-type islets were transplanted into Gcgr(-/-) or Gcgr(Hep)(-/-) mice. Wild-type islets beneath the renal capsule of Gcgr(-/-) or Gcgr(Hep)(-/-) mice exhibited an increased rate of α-cell proliferation and expansion of α-cell area, consistent with changes exhibited by endogenous α-cells in Gcgr(-/-) and Gcgr(Hep)(-/-) pancreata. These results suggest that a circulating factor generated after disruption of hepatic Gcgr signaling can increase α-cell proliferation independent of direct pancreatic input. Identification of novel factors regulating α-cell proliferation and mass may facilitate the generation and expansion of α-cells for transdifferentiation into β-cells and the treatment of diabetes.

Figures

FIG. 1.
FIG. 1.
Ablation of the hepatocyte Gcgr leads to hypoglycemia and improved glucose tolerance. A: Glycemia in 12-week-old male GcgrHep−/− and littermate controls fasted for 5 h or 16 h (n = 10–12 mice per group). B: Intraperitoneal (IP) glucose challenge in 9-week-old male GcgrHep−/− and littermate controls fasted for 16 h (n = 8–10 mice). Plasma glucagon (C) and insulin (D) levels measured before and 15 min after IP glucose injection (n = 8–10 mice per group). E: Insulin-to-glucose ratio before and 15 min after IP glucose injection. F: Oral glucose challenge in 8-week-old GcgrHep−/− males and littermate controls fasted for 16 h (n = 10–12 mice per group). Plasma glucagon (G) and insulin (H) levels measured before and 15 min after oral glucose administration (n = 10–12 mice per group). I: Insulin-to-glucose ratio before and 15 min after oral glucose injection. Data are mean ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001 vs. wild-type mice.
FIG. 2.
FIG. 2.
Ablation of the Gcgr in hepatocytes increases insulin sensitivity. A: Intraperitoneal (IP) insulin tolerance test in 13-week-old male GcgrHep−/− and littermate controls fasted for 5 h (n = 8–10 mice). B: Area under the curve (AUC) glucose from the IP insulin tolerance test shown in A. Hyperinsulinemic euglycemic clamp performed in conscious Gcgr−/− (C, E) or GcgrHep−/− (E, F) males and littermate controls fasted for 5 h (n = 5–7 mice). C and E: Glycemic excursion during stabilization and steady-state phase. D and F: Glucose infusion rates during steady state to maintain euglycemia. Data are mean ± SEM. *P < 0.05, **P < 0.01 vs. wild-type mice.
FIG. 3.
FIG. 3.
Whole-body ablation of the Gcgr increases pancreatic weight, α-cell mass, and α-cell proliferation. A: Histological sections stained for insulin (left panels) or glucagon (right panels). B: Pancreas weight of 20-week-old Gcgr−/− males and littermate controls corrected for body weight (n = 8–10 mice). Pancreatic β-cell mass (C) and α-cell mass (D) per gram of pancreas. Data are mean ± SEM. ***P < 0.001 vs. Gcgr+/+. Representative immunofluorescent sections stained for dapi (blue), glucagon (green), and Ki67 (red) from 22-week-old Gcgr−/− mice (F) and littermate controls (E). F′: Higher magnification of Ki67-positive nuclei from F. White arrows indicate Ki67-positive nuclei of α-cells. G: The α-cell proliferation rate (percentage of glucagon+/Ki67+ cells per total glucagon+ cells, n = 4 per group; 3 depths/pancreas). Data are mean ± SEM. *P < 0.05 vs. Gcgr+/+.
FIG. 4.
FIG. 4.
Ablation of the Gcgr in hepatocytes results in increased pancreas weight and α-cell hyperplasia. A: Representative histological sections stained for insulin (left panels) or glucagon (right panels). B: Pancreas weight of 20-week-old GcgrHep−/− males and littermate controls corrected for body weight (n = 8–10 mice). Pancreatic β-cell mass (C) and α-cell mass (D) per gram of pancreas. Data are mean ± SEM. ***P < 0.001 vs. wild type. Representative immunofluorescent sections stained for dapi (blue), glucagon (green), and Ki67 (red) from 22-week-old GcgrHep−/− mice (F) and littermate controls (E). E′ and F′: Higher magnification of sections containing Ki67-positive nuclei, denoted by white arrows, in α-cells. Insets are magnifications of selected Ki67-positive nuclei of α-cells (white box). White scale bar represents 50 μm. G: The α-cell proliferation rate (percentage of glucagon+/Ki67+ cells per total glucagon+ cells, n = 4 per group; 3 depths/pancreas). Data are mean ± SEM. ***P < 0.001 vs. Gcgrflox.
FIG. 5.
FIG. 5.
Gcgr+/+ islets transplanted into Gcgr−/− mice have development of α-cell hyperplasia. A: Experimental schematic showing islets from 14-week-old Gcgr+/+ or Gcgr−/− mice transplanted beneath the right and left kidney capsule of 14-week-old Gcgr+/+ or Gcgr−/− mice, respectively. The kidneys containing the islet graft were removed for analysis 8 weeks later. B: Hormone content of freshly isolated islets of 14-week-old Gcgr+/+ (open bars) and Gcgr−/− (closed bars) mice. Data are mean ± SEM. *P < 0.05, ***P < 0.001 vs. Gcgr+/+ islets. C: Hormone content of Gcgr+/+ grafts removed from 22-week-old mice Gcgr+/+ (open bars) and Gcgr−/− (light grey bars) recipient mice 8 weeks after transplant. Data are mean ± SEM. *P < 0.05, **P < 0.01 vs. Gcgr+/+ recipients. D: Hormone content of Gcgr−/− grafts removed from 22-week-old mice Gcgr+/+ (closed bars) and Gcgr−/− (dark grey bars) recipient mice 8 weeks after transplantation. Data are mean ± SEM. *P < 0.05, ***P < 0.001 vs. Gcgr−/− recipients. Representative islet graft sections from Gcgr+/+ (donor) to Gcgr+/+ (recipient; E) and Gcgr+/+ (donor) to Gcgr−/− (recipient; F) mice stained for insulin (green) and glucagon (red). E′ and F′: Insets of E and F (white box). Representative islet graft sections from Gcgr−/− (donor) to Gcgr+/+ (recipient; G) and Gcgr−/− (donor) to Gcgr−/− (recipient; H) mice stained for insulin (green) and glucagon (red). G′ and H′: Insets of G and H (white box). White scale bar represents 50 μm. Percentage of islet grafts that stained positive for glucagon for Gcgr+/+ donor (I) and Gcgr−/− donor islets (J) into Gcgr+/+ and Gcgr−/− mice (n = 3/mice, 3 sections/islet graft). Percent glucagon area is defined as the percentage of glucagon area/total insulin + glucagon area. Data are mean ± SEM. ***P < 0.001 vs. Gcgr+/+ recipient grafts.
FIG. 6.
FIG. 6.
Gcgr+/+ islets transplanted into Gcgr−/− recipients exhibit increased α-cell proliferation. Islet graft sections from Gcgr+/+ (donor) to Gcgr+/+ (recipient; A) and Gcgr+/+ (donor) to Gcgr−/− (recipient; B) mice stained for Ki67 (red), glucagon (green), and dapi (blue). A′ and B′: Insets of A and B (white box from A, B). White arrows indicate Ki67-positive α-cells. Islet graft sections from Gcgr−/− (donor) to Gcgr+/+ (recipient; C) and Gcgr−/− (donor) to Gcgr−/− (recipient; D) mice stained for Ki67 (red), glucagon (green), and dapi (blue). C′ and D′: Insets of C and D (white box from C, D). White arrows indicate Ki67-positive α-cells. White scale bar represents 50 μm. Proliferation rate of α-cells in grafts of islets from 14-week-old Gcgr+/+ (E) or Gcgr−/− (F) mice (donors) transplanted beneath the right and left kidney capsule, respectively, of 14-week-old Gcgr+/+ (recipient) or Gcgr−/− (recipient) mice. The kidneys containing the islet graft were removed for analysis 8 weeks later (n = 3/mice, 3 sections/islet graft). Data are mean ± SEM. **P < 0.01 vs. control grafts (donor and recipient of same genotype). G: Proliferation rate of α-cells in grafts of islets from 14-week-old Gcgr+/+ mice transplanted beneath the right and left kidney capsule of 14-week-old Gcgr+/+ or Gcgr−/− mice. The kidney containing the islet graft was removed for analysis 1 week later (n = 4–6/mice, 3 sections/graft). Data are mean ± SEM. *P < 0.05 vs. Gcgr+/+ recipient grafts.
FIG. 7.
FIG. 7.
Wild-type islets transplanted into GcgrHep−/− recipients for 4 weeks have increased α-cell area and proliferation. Representative islet graft sections from Gcgr+/+ (donor) to Gcgrflox (recipient; A) and Gcgr+/+ (donor) to GcgrHep−/− (recipient; B) mice stained for insulin (green) and glucagon (red). A′ and B′: Insets of A and B (white box). C: Percentage of islet grafts that stained positive for glucagon for Gcgr+/+ donor islets into Gcgrflox (recipient, open bars) and GcgrHep−/− (recipient, closed bars) mice (n = 4 mice, 3 sections/islet graft). Percent glucagon area is defined as the percentage of glucagon area/total insulin + glucagon area. Data are mean ± SEM. *P < 0.05 vs. Gcgrflox recipient grafts. Islet grafts sections from Gcgr+/+ (donor) to Gcgrflox (recipient; D) and Gcgr+/+ (donor) to GcgrHep−/− (recipient; E) mice stained for Ki67 (red), glucagon (green), and dapi (blue). D′ and E′: Insets of D and E (white box). White arrows indicate the Ki67-positive α-cells. White scale bar represents 25 μm. F: Proliferation rate of α-cells in Gcgr+/+ islet graft (donor) transplanted into Gcgrflox (recipient, open bars) or GcgrHep−/− (recipient, closed bars) mice (n = 4 mice, 3 sections/islet graft). Data are mean ± SEM. **P < 0.01 vs. Gcgrflox recipient grafts.

References

    1. Seymour PA, Sander M. Historical perspective: beginnings of the beta-cell: current perspectives in beta-cell development. Diabetes 2011;60:364–376
    1. Bramswig NC, Kaestner KH. Transcriptional regulation of α-cell differentiation. Diabetes Obes Metab 2011;13(Suppl. 1):13–20
    1. Müller WA, Faloona GR, Aguilar-Parada E, Unger RH. Abnormal alpha-cell function in diabetes. Response to carbohydrate and protein ingestion. N Engl J Med 1970;283:109–115
    1. Dunning BE, Gerich JE. The role of alpha-cell dysregulation in fasting and postprandial hyperglycemia in type 2 diabetes and therapeutic implications. Endocr Rev 2007;28:253–283
    1. Dufrane D, Maillart JF, Aouassar N, Goebbels RM, Guiot Y, Gianello P. Native pancreatic alpha-cell adaptation in streptozotocin-induced diabetic primates: importance for pig islet xenotransplantation. Xenotransplantation 2009;16:152–163
    1. Yoon KH, Ko SH, Cho JH, et al. Selective beta-cell loss and alpha-cell expansion in patients with type 2 diabetes mellitus in Korea. J Clin Endocrinol Metab 2003;88:2300–2308
    1. Collombat P, Xu X, Ravassard P, et al. The ectopic expression of Pax4 in the mouse pancreas converts progenitor cells into alpha and subsequently beta cells. Cell 2009;138:449–462
    1. Thorel F, Népote V, Avril I, et al. Conversion of adult pancreatic alpha-cells to beta-cells after extreme beta-cell loss. Nature 2010;464:1149–1154
    1. Chung CH, Hao E, Piran R, Keinan E, Levine F. Pancreatic β-cell neogenesis by direct conversion from mature α-cells. Stem Cells 2010;28:1630–1638
    1. 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 USA 2003;100:1438–1443
    1. Vuguin PM, Kedees MH, Cui L, et al. Ablation of the glucagon receptor gene increases fetal lethality and produces alterations in islet development and maturation. Endocrinology 2006;147:3995–4006
    1. Chen M, Gavrilova O, Zhao WQ, 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
    1. Webb GC, Akbar MS, Zhao C, Swift HH, Steiner DF. Glucagon replacement via micro-osmotic pump corrects hypoglycemia and alpha-cell hyperplasia in prohormone convertase 2 knockout mice. Diabetes 2002;51:398–405
    1. Sloop KW, Cao JX, Siesky AM, et al. Hepatic and glucagon-like peptide-1-mediated reversal of diabetes by glucagon receptor antisense oligonucleotide inhibitors. J Clin Invest 2004;113:1571–1581
    1. Ali S, Lamont BJ, Charron MJ, Drucker DJ. Dual elimination of the glucagon and GLP-1 receptors in mice reveals plasticity in the incretin axis. J Clin Invest 2011;121:1917–1929
    1. Chen M, Mema E, Kelleher J, et al. Absence of the glucagon-like peptide-1 receptor does not affect the metabolic phenotype of mice with liver-specific G(s)α deficiency. Endocrinology 2011;152:3343–3350
    1. Postic C, Magnuson MA. DNA excision in liver by an albumin-Cre transgene occurs progressively with age. Genesis 2000;26:149–150
    1. Flock G, Baggio LL, Longuet C, Drucker DJ. Incretin receptors for glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide are essential for the sustained metabolic actions of vildagliptin in mice. Diabetes 2007;56:3006–3013
    1. Maida A, Hansotia T, Longuet C, Seino Y, Drucker DJ. Differential importance of glucose-dependent insulinotropic polypeptide vs glucagon-like peptide 1 receptor signaling for beta cell survival in mice. Gastroenterology 2009;137:2146–2157
    1. Brissova M, Shostak A, Shiota M, et al. Pancreatic islet production of vascular endothelial growth factor—a is essential for islet vascularization, revascularization, and function. Diabetes 2006;55:2974–2985
    1. Ayala JE, Bracy DP, McGuinness OP, Wasserman DH. Considerations in the design of hyperinsulinemic-euglycemic clamps in the conscious mouse. Diabetes 2006;55:390–397
    1. Brissova M, Fowler M, Wiebe P, et al. Intraislet endothelial cells contribute to revascularization of transplanted pancreatic islets. Diabetes 2004;53:1318–1325
    1. Fujita Y, Wideman RD, Asadi A, et al. Glucose-dependent insulinotropic polypeptide is expressed in pancreatic islet alpha-cells and promotes insulin secretion. Gastroenterology 2010;138:1966–1975
    1. Korsgren O, Andersson A, Jansson L, Sundler F. Reinnervation of syngeneic mouse pancreatic islets transplanted into renal subcapsular space. Diabetes 1992;41:130–135
    1. Imai J, Katagiri H, Yamada T, et al. Regulation of pancreatic beta cell mass by neuronal signals from the liver. Science 2008;322:1250–1254
    1. Ellingsgaard H, Ehses JA, Hammar EB, et al. Interleukin-6 regulates pancreatic alpha-cell mass expansion. Proc Natl Acad Sci USA 2008;105:13163–13168
    1. Liu Z, Stanojevic V, Avadhani S, Yano T, Habener JF. Stromal cell-derived factor-1 (SDF-1)/chemokine (C-X-C motif) receptor 4 (CXCR4) axis activation induces intra-islet glucagon-like peptide-1 (GLP-1) production and enhances beta cell survival. Diabetologia 2011;54:2067–2076
    1. 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.
    1. Gu W, Yan H, Winters KA, et al. Long-term inhibition of the glucagon receptor with a monoclonal antibody in mice causes sustained improvement in glycemic control, with reversible alpha-cell hyperplasia and hyperglucagonemia. J Pharmacol Exp Ther 2009;331:871–881
    1. Petersson B, Hellman B. Effects of long term administration of glucagon on the pancreatic islet tissue of rats and guinea-pigs. Acta Endocrinol (Copenh) 1963;44:139–149
    1. 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
    1. Ehrlich P, Tucker D, Asa SL, Brubaker PL, Drucker DJ. Inhibition of pancreatic proglucagon gene expression in mice bearing subcutaneous endocrine tumors. Am J Physiol 1994;267:E662–E671
    1. Xie T, Chen M, Weinstein LS. Pancreas-specific Gsalpha deficiency has divergent effects on pancreatic alpha- and beta-cell proliferation. J Endocrinol 2010;206:261–269
    1. Xie T, Chen M, Zhang QH, Ma Z, Weinstein LS. Beta cell-specific deficiency of the stimulatory G protein alpha-subunit Gsalpha leads to reduced beta cell mass and insulin-deficient diabetes. Proc Natl Acad Sci USA 2007;104:19601–19606
    1. Liu Z, Kim W, Chen Z, et al. Insulin and glucagon regulate pancreatic α-cell proliferation. PLoS ONE 2011;6:e16096.
    1. Winzell MS, Brand CL, Wierup N, et al. Glucagon receptor antagonism improves islet function in mice with insulin resistance induced by a high-fat diet. Diabetologia 2007;50:1453–1462
    1. Kellendonk C, Opherk C, Anlag K, Schütz G, Tronche F. Hepatocyte-specific expression of Cre recombinase. Genesis 2000;26:151–153
    1. Chen L, Komiya I, Inman L, et al. Effects of hypoglycemia and prolonged fasting on insulin and glucagon gene expression. Studies with in situ hybridization. J Clin Invest 1989;84:711–714
    1. Shigeyama Y, Kobayashi T, Kido Y, et al. Biphasic response of pancreatic beta-cell mass to ablation of tuberous sclerosis complex 2 in mice. Mol Cell Biol 2008;28:2971–2979
    1. Sund NJ, Vatamaniuk MZ, Casey M, et al. Tissue-specific deletion of Foxa2 in pancreatic beta cells results in hyperinsulinemic hypoglycemia. Genes Dev 2001;15:1706–1715
    1. Kawamori D, Akiyama M, Hu J, Hambro B, Kulkarni RN. Growth factor signalling in the regulation of α-cell fate. Diabetes Obes Metab 2011;13(Suppl. 1):21–30
    1. Flier SN, Kulkarni RN, Kahn CR. Evidence for a circulating islet cell growth factor in insulin-resistant states. Proc Natl Acad Sci USA 2001;98:7475–7480
    1. Gu W, Winters KA, Motani AS, et al. Glucagon receptor antagonist-mediated improvements in glycemic control are dependent on functional pancreatic GLP-1 receptor. Am J Physiol Endocrinol Metab 2010;299:E624–E632
    1. Zhou C, Dhall D, Nissen NN, Chen CR, 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
    1. Henopp T, Anlauf M, Schmitt A, et al. Glucagon cell adenomatosis: a newly recognized disease of the endocrine pancreas. J Clin Endocrinol Metab 2009;94:213–217
    1. Willcox A, Richardson SJ, Bone AJ, Foulis AK, Morgan NG. Evidence of increased islet cell proliferation in patients with recent-onset type 1 diabetes. Diabetologia 2010;53:2020–2028

Source: PubMed

3
Abonnere