Chronic administration of the glucagon-like peptide-1 analog, liraglutide, delays the onset of diabetes and lowers triglycerides in UCD-T2DM rats

Bethany P Cummings, Kimber L Stanhope, James L Graham, Denis G Baskin, Steven C Griffen, Cecilia Nilsson, Anette Sams, Lotte B Knudsen, Kirsten Raun, Peter J Havel, Bethany P Cummings, Kimber L Stanhope, James L Graham, Denis G Baskin, Steven C Griffen, Cecilia Nilsson, Anette Sams, Lotte B Knudsen, Kirsten Raun, Peter J Havel

Abstract

Objective: The efficacy of liraglutide, a human glucagon-like peptide-1 (GLP-1) analog, to prevent or delay diabetes in UCD-T2DM rats, a model of polygenic obese type 2 diabetes, was investigated.

Research design and methods: At 2 months of age, male rats were divided into three groups: control, food-restricted, and liraglutide. Animals received liraglutide (0.2 mg/kg s.c.) or vehicle injections twice daily. Restricted rats were food restricted to equalize body weights to liraglutide-treated rats. Half of the animals were followed until diabetes onset, whereas the other half of the animals were killed at 6.5 months of age for tissue collection.

Results: Before diabetes onset energy intake, body weight, adiposity, and liver triglyceride content were higher in control animals compared with restricted and liraglutide-treated rats. Energy-restricted animals had lower food intake than liraglutide-treated animals to maintain the same body weights, suggesting that liraglutide increases energy expenditure. Liraglutide treatment delayed diabetes onset by 4.1 ± 0.8 months compared with control (P < 0.0001) and by 1.3 ± 0.8 months compared with restricted animals (P < 0.05). Up to 6 months of age, energy restriction and liraglutide treatment lowered fasting plasma glucose and A1C concentrations compared with control animals. In contrast, liraglutide-treated animals exhibited lower fasting plasma insulin, glucagon, and triglycerides compared with both control and restricted animals. Furthermore, energy-restricted and liraglutide-treated animals exhibited more normal islet morphology.

Conclusions: Liraglutide treatment delays the development of diabetes in UCD-T2DM rats by reducing energy intake and body weight, and by improving insulin sensitivity, improving lipid profiles, and maintaining islet morphology.

Figures

FIG. 1.
FIG. 1.
Kaplan-Meier analysis of diabetes incidence in control, restricted, and liraglutide-treated animals up to 6 months (n = 32 per group) (A) and 12 months (n = 16 per group) (B) of age. ***P < 0.0001, **P < 0.01 compared with control; *P < 0.05 compared with the food-restricted group by log-rank test. Nonfasting blood glucose in control, restricted, and liraglutide-treated animals (C). ***P < 0.0001 by two-factor (time and treatment) repeated-measures ANOVA, ***P < 0.001 compared with restricted and liraglutide-treated animals by Bonferroni post-test. Control: n = 27; restricted: n = 31; liraglutide-treated animals: n = 32.
FIG. 2.
FIG. 2.
Energy intake (A) and body weight (B) in control, restricted, and liraglutide-treated animals. ***P < 0.0001 by two-factor (time and treatment) repeated-measures ANOVA; ***P < 0.001 compared with restricted and liraglutide-treated animals; *P < 0.05 liraglutide-treated animals versus restricted by Bonferroni post-test. Control: n = 27; restricted: n = 31; liraglutide-treated animals: n = 32.
FIG. 3.
FIG. 3.
Fasting circulating glucose (A), A1C (B), and insulin (C) in control, restricted, and liraglutide-treated animals. ***P < 0.0001 by two-factor (time and treatment) repeated-measures ANOVA; ***P < 0.001, *P < 0.05 compared with restricted and liraglutide-treated animals; **P < 0.001 compared with restricted and control; ++P < 0.001 compared with restricted; +P < 0.05 compared with control by Bonferroni post-test. Control: n = 27; restricted: n = 31; liraglutide-treated animals: n = 32.
FIG. 4.
FIG. 4.
Fasting plasma TG (A), cholesterol (B), glucagon (C), and adiponectin (D) in control, food-restricted, and liraglutide-treated animals. ***P < 0.0001, *P < 0.05 by two-factor (time and treatment) repeated-measures ANOVA; ***P < 0.001 compared with food-restricted and control; **P < 0.001 compared with food-restricted; *P < 0.05 compared with control; +P < 0.05 compared with food-restricted; ++P < 0.05 compared with food-restricted and liraglutide-treated animals by Bonferroni post-test. Control: n = 27; restricted: n = 31; liraglutide-treated animals: n = 32.
FIG. 5.
FIG. 5.
Representative pancreas sections from control, food-restricted, and liraglutide-treated animals at 6 months of age. Hematoxylin and eosin staining for control (A), restricted (D), and liraglutide-treated animals (G) is shown. Anti-insulin immunostaining for control (B), restricted (E), and liraglutide-treated animals (H) is shown. Anti-glucagon immunostaining for control (C), restricted (F), and liraglutide-treated animals (I) is shown. (A high-quality digital representation of this figure is available in the online issue.)

References

    1. Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007;132:2131–2157
    1. Herman GA, Stein PP, Thornberry NA, Wagner JA. Dipeptidyl peptidase-4 inhibitors for the treatment of type 2 diabetes: focus on sitagliptin. Clin Pharmacol Ther 2007;81:761–767
    1. Kreymann B, Williams G, Ghatei MA, Bloom SR. Glucagon-like peptide-1 7–36: a physiological incretin in man. Lancet 1987;2:1300–1304
    1. Mojsov S, Weir GC, Habener JF. Insulinotropin: glucagon-like peptide I (7-37) co-encoded in the glucagon gene is a potent stimulator of insulin release in the perfused rat pancreas. J Clin Invest 1987;79:616–619
    1. Kim Chung le T, Hosaka T, Yoshida M, Harada N, Sakaue H, Sakai T, Nakaya Y. Exendin-4, a GLP-1 receptor agonist, directly induces adiponectin expression through protein kinase A pathway and prevents inflammatory adipokine expression. Biochem Biophys Res Commun 2009;390:613–618
    1. Flint A, Raben A, Astrup A, Holst JJ. Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans. J Clin Invest 1998;101:515–520
    1. Gutzwiller JP, Drewe J, Goke B, Schmidt H, Rohrer B, Lareida J, Beglinger C. Glucagon-like peptide-1 promotes satiety and reduces food intake in patients with diabetes mellitus type 2. Am J Physiol 1999;276:R1541–R1544
    1. Wren AM, Bloom SR. Gut hormones and appetite control. Gastroenterology 2007;132:2116–2130
    1. D'Alessio DA, Vahl TP. Glucagon-like peptide 1: evolution of an incretin into a treatment for diabetes. Am J Physiol Endocrinol Metab 2004;286:E882–E890
    1. Merani S, Truong W, Emamaullee JA, Toso C, Knudsen LB, Shapiro AM. Liraglutide, a long-acting human glucagon-like peptide 1 analog, improves glucose homeostasis in marginal mass islet transplantation in mice. Endocrinology 2008;149:4322–4328
    1. Salehi M, Aulinger BA, D'Alessio DA. Targeting beta-cell mass in type 2 diabetes: promise and limitations of new drugs based on incretins. Endocr Rev 2008;29:367–379
    1. D'Alessio DA, Kahn SE, Leusner CR, Ensinck JW. Glucagon-like peptide 1 enhances glucose tolerance both by stimulation of insulin release and by increasing insulin-independent glucose disposal. J Clin Invest 1994;93:2263–2266
    1. Gedulin BR, Nikoulina SE, Smith PA, Gedulin G, Nielsen LL, Baron AD, Parkes DG, Young AA. Exenatide (exendin-4) improves insulin sensitivity and β-cell mass in insulin-resistant obese fa/fa Zucker rats independent of glycemia and body weight. Endocrinology 2005;146:2069–2076
    1. Gonzalez N, Acitores A, Sancho V, Valverde I, Villanueva-Penacarrillo ML. Effect of GLP-1 on glucose transport and its cell signalling in human myocytes. Regul Pept 2005;126:203–211
    1. Prigeon RL, Quddusi S, Paty B, D'Alessio DA. Suppression of glucose production by GLP-1 independent of islet hormones: a novel extrapancreatic effect. Am J Physiol Endocrinol Metab 2003;285:E701–E707
    1. Redondo A, Trigo MV, Acitores A, Valverde I, Villanueva-Penacarrillo ML. Cell signalling of the GLP-1 action in rat liver. Mol Cell Endocrinol 2003;204:43–50
    1. Deacon CF, Johnsen AH, Holst JJ. Degradation of glucagon-like peptide-1 by human plasma in vitro yields an N-terminally truncated peptide that is a major endogenous metabolite in vivo. J Clin Endocrinol Metab 1995;80:952–957
    1. Agerso H, Jensen LB, Elbrond B, Rolan P, Zdravkovic M. The pharmacokinetics, pharmacodynamics, safety and tolerability of NN2211, a new long-acting GLP-1 derivative, in healthy men. Diabetologia 2002;45:195–202
    1. Knudsen LB, Nielsen PF, Huusfeldt PO, Johansen NL, Madsen K, Pedersen FZ, Thogersen H, Wilken M, Agerso H. Potent derivatives of glucagon-like peptide-1 with pharmacokinetic properties suitable for once daily administration. J Med Chem 2000;43:1664–1669
    1. Buse JB, Rosenstock J, Sesti G, Schmidt WE, Montanya E, Brett JH, Zychma M, Blonde L. Liraglutide once a day versus exenatide twice a day for type 2 diabetes: a 26-week randomised, parallel-group, multinational, open-label trial (LEAD-6). Lancet 2009;374:39–47
    1. Garber A, Henry R, Ratner R, Garcia-Hernandez PA, Rodriguez-Pattzi H, Olvera-Alvarez I, Hale PM, Zdravkovic M, Bode B. Liraglutide versus glimepiride monotherapy for type 2 diabetes (LEAD-3 Mono): a randomised, 52-week, phase III, double-blind, parallel-treatment trial. Lancet 2009;373:473–481
    1. Marre M, Shaw J, Brandle M, Bebakar WM, Kamaruddin NA, Strand J, Zdravkovic M, Le Thi TD, Colagiuri S. Liraglutide, a once-daily human GLP-1 analogue, added to a sulphonylurea over 26 weeks produces greater improvements in glycaemic and weight control compared with adding rosiglitazone or placebo in subjects with type 2 diabetes (LEAD-1 SU). Diabet Med 2009;26:268–278
    1. Nauck M, Frid A, Hermansen K, Shah NS, Tankova T, Mitha IH, Zdravkovic M, During M, Matthews DR. Efficacy and safety comparison of liraglutide, glimepiride, and placebo, all in combination with metformin, in type 2 diabetes: the LEAD (liraglutide effect and action in diabetes)-2 study. Diabetes Care 2009;32:84–90
    1. Russell-Jones D, Vaag A, Schmitz O, Sethi BK, Lalic N, Antic S, Zdravkovic M, Ravn GM, Simo R. Liraglutide vs insulin glargine and placebo in combination with metformin and sulfonylurea therapy in type 2 diabetes mellitus (LEAD-5 met+SU): a randomised controlled trial. Diabetologia 2009;52:2046–2055
    1. Zinman B, Gerich J, Buse JB, Lewin A, Schwartz S, Raskin P, Hale PM, Zdravkovic M, Blonde L. Efficacy and safety of the human glucagon-like peptide-1 analog liraglutide in combination with metformin and thiazolidinedione in patients with type 2 diabetes (LEAD-4 Met+TZD). Diabetes Care 2009;32:1224–1230
    1. Cummings BP, Digitale EK, Stanhope KL, Graham JL, Baskin DG, Reed BJ, Sweet IR, Griffen SC, Havel PJ. Development and characterization of a novel rat model of type 2 diabetes mellitus: the UC Davis type 2 diabetes mellitus UCD-T2DM rat. Am J Physiol Regul Integr Comp Physiol 2008;295:R1782–R1793
    1. Bergeron R, Yao J, Woods JW, Zycband EI, Liu C, Li Z, Adams A, Berger JP, Zhang BB, Moller DE, Doebber TW. Peroxisome proliferator-activated receptor (PPAR)-α agonism prevents the onset of type 2 diabetes in Zucker diabetic fatty rats: a comparison with PPARγ agonism. Endocrinology 2006;147:4252–4262
    1. Kessler MA, Meinitzer A, Petek W, Wolfbeis OS. Microalbuminuria and borderline-increased albumin excretion determined with a centrifugal analyzer and the Albumin Blue 580 fluorescence assay. Clin Chem 1997;43:996–1002
    1. Folch J, Lees M, Sloane Stanley GH. A simple method for the isolation and purification of total lipides from animal tissues. J Biol Chem 1957;226:497–509
    1. Bell GE, Stern JS. Evaluation of body composition of young obese and lean Zucker rats. Growth 1977;41:63–80
    1. Cummings BP, Stanhope KL, Graham JL, Evans JL, Baskin DG, Griffen SC, Havel PJ. Dietary fructose accelerates the development of diabetes in UCD-T2DM rats: amelioration by the antioxidant, α-lipoic acid. Am J Physiol Regul Integr Comp Physiol 2010;298:R1333–R1342
    1. Larsen PJ, Fledelius C, Knudsen LB, Tang-Christensen M. Systemic administration of the long-acting GLP-1 derivative NN2211 induces lasting and reversible weight loss in both normal and obese rats. Diabetes 2001;50:2530–2539
    1. Raun K, von Voss P, Gotfredsen CF, Golozoubova V, Rolin B, Knudsen LB. Liraglutide, a long-acting glucagon-like peptide-1 analog, reduces body weight and food intake in obese candy-fed rats, whereas a dipeptidyl peptidase-IV inhibitor, vildagliptin, does not. Diabetes 2007;56:8–15
    1. Kanaya AM, Narayan KM. Prevention of type 2 diabetes: data from recent trials. Prim Care 2003;30:511–526
    1. Abu-Hamdah R, Rabiee A, Meneilly GS, Shannon RP, Andersen DK, Elahi D. Clinical review: the extrapancreatic effects of glucagon-like peptide-1 and related peptides. J Clin Endocrinol Metab 2009;94:1843–1852
    1. Vahl TP, Tauchi M, Durler TS, Elfers EE, Fernandes TM, Bitner RD, Ellis KS, Woods SC, Seeley RJ, Herman JP, D'Alessio DA. Glucagon-like peptide-1 (GLP-1) receptors expressed on nerve terminals in the portal vein mediate the effects of endogenous GLP-1 on glucose tolerance in rats. Endocrinology 2007;148:4965–4973
    1. Morino K, Petersen KF, Shulman GI. Molecular mechanisms of insulin resistance in humans and their potential links with mitochondrial dysfunction. Diabetes 2006;55(Suppl. 2):S9–S15
    1. Dunning BE, Gerich JE. The role of α-cell dysregulation in fasting and postprandial hyperglycemia in type 2 diabetes and therapeutic implications. Endocr Rev 2007;28:253–283
    1. Ayala JE, Bracy DP, James FD, Julien BM, Wasserman DH, Drucker DJ. The glucagon-like peptide-1 receptor regulates endogenous glucose production and muscle glucose uptake independent of its incretin action. Endocrinology 2009;150:1155–1164
    1. Mueller WM, Gregoire FM, Stanhope KL, Mobbs CV, Mizuno TM, Warden CH, Stern JS, Havel PJ. Evidence that glucose metabolism regulates leptin secretion from cultured rat adipocytes. Endocrinology 1998;139:551–558
    1. Havel PJ. Update on adipocyte hormones: regulation of energy balance and carbohydrate/lipid metabolism. Diabetes 2004;53:S143–S148
    1. Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC. β-Cell deficit and increased β-cell apoptosis in humans with type 2 diabetes. Diabetes 2003;52:102–110

Source: PubMed

3
S'abonner