GLP-1R agonist liraglutide activates cytoprotective pathways and improves outcomes after experimental myocardial infarction in mice

Mohammad Hossein Noyan-Ashraf, M Abdul Momen, Kiwon Ban, Al-Muktafi Sadi, Yu-Qing Zhou, Ali M Riazi, Laurie L Baggio, R Mark Henkelman, Mansoor Husain, Daniel J Drucker, Mohammad Hossein Noyan-Ashraf, M Abdul Momen, Kiwon Ban, Al-Muktafi Sadi, Yu-Qing Zhou, Ali M Riazi, Laurie L Baggio, R Mark Henkelman, Mansoor Husain, Daniel J Drucker

Abstract

Objective: Glucagon-like peptide-1 receptor (GLP-1R) agonists are used to treat type 2 diabetes, and transient GLP-1 administration improved cardiac function in humans after acute myocardial infarction (MI) and percutaneous revascularization. However, the consequences of GLP-1R activation before ischemic myocardial injury remain unclear.

Research design and methods: We assessed the pathophysiology and outcome of coronary artery occlusion in normal and diabetic mice pretreated with the GLP-1R agonist liraglutide.

Results: Male C57BL/6 mice were treated twice daily for 7 days with liraglutide or saline followed by induction of MI. Survival was significantly higher in liraglutide-treated mice. Liraglutide reduced cardiac rupture (12 of 60 versus 46 of 60; P = 0.0001) and infarct size (21 +/- 2% versus 29 +/- 3%, P = 0.02) and improved cardiac output (12.4 +/- 0.6 versus 9.7 +/- 0.6 ml/min; P = 0.002). Liraglutide also modulated the expression and activity of cardioprotective genes in the mouse heart, including Akt, GSK3beta, PPARbeta-delta, Nrf-2, and HO-1. The effects of liraglutide on survival were independent of weight loss. Moreover, liraglutide conferred cardioprotection and survival advantages over metformin, despite equivalent glycemic control, in diabetic mice with experimental MI. The cardioprotective effects of liraglutide remained detectable 4 days after cessation of therapy and may be partly direct, because liraglutide increased cyclic AMP formation and reduced the extent of caspase-3 activation in cardiomyocytes in a GLP-1R-dependent manner in vitro.

Conclusions: These findings demonstrate that GLP-1R activation engages prosurvival pathways in the normal and diabetic mouse heart, leading to improved outcomes and enhanced survival after MI in vivo.

Figures

FIG. 1.
FIG. 1.
Liraglutide pretreatment improves outcomes after MI in mice. Kaplan-Meier survival curves show survival after MI in nondiabetic mice (A): sham (n = 20), liraglutide-MI (75 μg/kg, n = 35 or 200 μg/kg, n = 60), PBS-MI (n = 60), and pair-fed mice (n = 25), P = 0.0001 for LIR 75 and 200 versus PBS; and diabetic mice (B): sham (n = 15, 5/treatment group), PBS-MI (n = 18), metformin-MI (n = 18), liraglutide-MI (75 μg/kg, n = 18), P = 0.04 for LIR 75 versus PBS. C: Frequency and timing of cardiac rupture in nondiabetic mice is shown as a percentage of total group. One-week pretreatment with liraglutide (200 or 75 μg/kg i.p. twice daily) had no significant effects on random blood glucose levels in adult nondiabetic mice (PBS: 7.6 ± 4.0 versus LIR 200: 6.3 ± 0.6 versus LIR 75: 6.1 ± 0.4 mmol/l; P = 0.72). Blood glucose levels in diabetic mice are shown in supplemental Table 1 (available in the online appendix).
FIG. 2.
FIG. 2.
Effects of liraglutide pretreatment on infarct size and heart weight. A: Representative photomicrographs of H-E–stained hearts 28 days post-MI depict decreased infarct size (arrows) in liraglutide- (n = 36) versus PBS-treated mice (n = 21) as confirmed by morphometric quantification of percent total LV circumference (*P = 0.025). B: HW/BW ratio was reduced in liraglutide- versus PBS-treated mice 28 days post-MI (n = 36 and 21, respectively; **P = 0.001). Data shown are means ± SE. (A high-quality digital representation of this figure is available in the online issue.)
FIG. 3.
FIG. 3.
Effects of liraglutide pretreatment on levels of cardiac genes and proteins. The expression of genes/proteins before left anterior descending ligation is analyzed in liraglutide-treated hearts. A: Representative Western blots for wild-type mice treated with LIR 200 as outlined in supplemental Fig. 1 (available in the online appendix). Corresponding densitometric quantification (n = 6/group) of fold changes in phosphorylation of prosurvival kinases Akt and GSK3β and in expression of PPAR-β/δ, Nrf2, and HO-1 are shown in the top five panels. The bottom panel depicts a representative agarose gel (n = 6/group) showing HO-1–specific mRNA levels by RT-PCR. Data shown are means ± SE; *P < 0.05. B–D: Representative Western blot analysis of cardiac prosurvival kinases in LIR 75–treated wild-type (B), LIR 75–treated Glp1r+/+ littermate controls (C), and LIR 75–treated Glp1r-/- (D) mice, respectively.
FIG. 4.
FIG. 4.
Effects of liraglutide pretreatment on cardioprotective signaling pathways after MI in mice. Liraglutide pretreatment for 7 days has persistent cardioprotective effects detectable 4 days post-MI as indicated by representative Western blots and corresponding densitometric quantification (n = 6/group) of fold changes in phosphorylation of Akt and GSK3β, cleavage of caspase-3, and expression of ANP and activity of MMP-9 by zymography in liraglutide- versus PBS-treated mice. Data shown are means ± SE; *P < 0.05.
FIG. 5.
FIG. 5.
Effects of liraglutide on recovery of LV function after I/R injury in isolated hearts. A: Effect of direct infusions of liraglutide- (0.3, 3, and 30 nmol/l, n = 5/group) or no treatment (vehicle, n = 21) either pre- or postischemia in isolated murine hearts subjected to experimental I/R ex vivo. B: Two additional groups of mice (n = 5/group) received i.p. injections of liraglutide b.i.d. for 1 or 7 days before ex vivo experiments. Data shown are means ± SE; *P < 0.01 compared with untreated controls.
FIG. 6.
FIG. 6.
Liraglutide induces cAMP formation and reduces caspase-3 activation in murine cardiomyocytes in vitro. A: Liraglutide (100 nmol/l) increases cAMP formation in cultured neonatal cardiomyocytes. The actions of liraglutide were abolished by the GLP-1R antagonist exendin9–39. ***P < 0.001. B: Liraglutide (L: 10–1,000 nmol/l) reduced TNF-α–induced activation of caspase-3 in a dose-dependent manner in cultured neonatal mouse cardiomyocytes. Cotreatment with exendin9–39 (Ex: 10 μmol/l) abolished the protective effects of liraglutide. Positive control represents treatment of cells with the potent apoptosis-inducing agent H2O2. Data shown are means ± SE; *P < 0.01 and **P < 0.001 versus cultures only treated with TNFα.

References

    1. Yellon DM, Dana A: The preconditioning phenomenon: a tool for the scientist or a clinical reality? Circ Res 87: 543– 550, 2000
    1. Drucker DJ, Nauck MA: The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368: 1696– 1705, 2006
    1. Ban K, Noyan-Ashraf MH, Hoefer J, Bolz SS, Drucker DJ, Husain M: Cardioprotective and vasodilatory actions of glucagon-like peptide 1 receptor are mediated through both glucagon-like peptide 1 receptor-dependent and -independent pathways. Circulation 117: 2340– 2350, 2008
    1. Vila Petroff MG, Egan JM, Wang X, Sollott SJ: Glucagon-like peptide-1 increases cAMP but fails to augment contraction in adult rat cardiac myocytes. Circ Res 89: 445– 452, 2001
    1. Barragan JM, Eng J, Rodriguez R, Blazquez E: Neural contribution to the effect of glucagon-like peptide-1-(7–36) amide on arterial blood pressure in rats. Am J Physiol 277: E784– E791, 1999
    1. Yamamoto H, Kishi T, Lee CE, Choi BJ, Fang H, Hollenberg AN, Drucker DJ, Elmquist JK: Glucagon-like peptide-1-responsive catecholamine neurons in the area postrema link peripheral glucagon-like peptide-1 with central autonomic control sites. J Neurosci 23: 2939– 2946, 2003
    1. Yamamoto H, Lee CE, Marcus JN, Williams TD, Overton JM, Lopez ME, Hollenberg AN, Baggio L, Saper CB, Drucker DJ, Elmquist JK: Glucagon-like peptide-1 receptor stimulation increases blood pressure and heart rate and activates autonomic regulatory neurons. J Clin Invest 110: 43– 52, 2002
    1. Nikolaidis LA, Elahi D, Hentosz T, Doverspike A, Huerbin R, Zourelias L, Stolarski C, Shen YT, Shannon RP: Recombinant glucagon-like peptide-1 increases myocardial glucose uptake and improves left ventricular performance in conscious dogs with pacing-induced dilated cardiomyopathy. Circulation 110: 955– 961, 2004
    1. Bose AK, Mocanu MM, Carr RD, Brand CL, Yellon DM: Glucagon-like peptide-1 (GLP-1) can directly protect the heart against ischemia/reperfusion injury. Diabetes 54: 146– 151, 2005
    1. Bose AK, Mocanu MM, Carr RD, Yellon DM: Glucagon like peptide-1 is protective against myocardial ischemia/reperfusion injury when given either as a preconditioning mimetic or at reperfusion in an isolated rat heart model. Cardiovasc Drugs Ther 19: 9– 11, 2005
    1. Bose AK, Mocanu MM, Carr RD, Yellon DM: Myocardial ischaemia–reperfusion injury is attenuated by intact glucagon like peptide-1 (GLP-1) in the in vitro rat heart and may involve the p70s6K pathway. Cardiovasc Drugs Ther 21: 253– 256, 2007
    1. Nikolaidis LA, Doverspike A, Hentosz T, Zourelias L, Shen YT, Elahi D, Shannon RP: Glucagon-like peptide-1 limits myocardial stunning following brief coronary occlusion and reperfusion in conscious canines. J Pharmacol Exp Ther 312: 303– 308, 2005
    1. Zhao T, Parikh P, Bhashyam S, Bolukoglu H, Poornima I, Shen YT, Shannon RP: Direct effects of glucagon-like peptide-1 on myocardial contractility and glucose uptake in normal and postischemic isolated rat hearts. J Pharmacol Exp Ther 317: 1106– 1113, 2006
    1. Nikolaidis LA, Mankad S, Sokos GG, Miske G, Shah A, Elahi D, Shannon RP: Effects of glucagon-like peptide-1 in patients with acute myocardial infarction and left ventricular dysfunction after successful reperfusion. Circulation 109: 962– 965, 2004
    1. Sokos GG, Nikolaidis LA, Mankad S, Elahi D, Shannon RP: Glucagon-like peptide-1 infusion improves left ventricular ejection fraction and functional status in patients with chronic heart failure. J Card Fail 12: 694– 699, 2006
    1. Sonne DP, Engstrom T, Treiman M: Protective effects of GLP-1 analogues exendin-4 and GLP-1(9–36) amide against ischemia-reperfusion injury in rat heart. Regul Pept 146: 243– 249, 2008
    1. Degn KB, Juhl CB, Sturis J, Jakobsen G, Brock B, Chandramouli V, Rungby J, Landau BR, Schmitz O: One week's treatment with the long-acting glucagon-like peptide 1 derivative liraglutide (NN2211) markedly improves 24-h glycemia and alpha- and beta-cell function and reduces endogenous glucose release in patients with type 2 diabetes. Diabetes 53: 1187– 1194, 2004
    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 50: 2530– 2539, 2001
    1. Vilsboll T, Zdravkovic M, Le-Thi T, Krarup T, Schmitz O, Courreges JP, Verhoeven R, Buganova I, Madsbad S: Liraglutide, a long-acting human GLP-1 Analog, given as monotherapy significantly improves glycemic control and lowers body weight without risk of hypoglycemia in patients with type 2 diabetes mellitus. Diabetes Care 6: 1608– 1610, 2007
    1. Ohta K, Nakajima T, Cheah AY, Zaidi SH, Kaviani N, Dawood F, You XM, Liu P, Husain M, Rabinovitch M: Elafin-overexpressing mice have improved cardiac function after myocardial infarction. Am J Physiol Heart Circ Physiol 287: H286– H292, 2004
    1. Yang Z, Berr SS, Gilson WD, Toufektsian MC, French BA: Simultaneous evaluation of infarct size and cardiac function in intact mice by contrast-enhanced cardiac magnetic resonance imaging reveals contractile dysfunction in noninfarcted regions early after myocardial infarction. Circulation 109: 1161– 1167, 2004
    1. Sun M, Chen M, Dawood F, Zurawska U, Li JY, Parker T, Kassiri Z, Kirshenbaum LA, Arnold M, Khokha R, Liu PP: Tumor necrosis factor-alpha mediates cardiac remodeling and ventricular dysfunction after pressure overload state. Circulation 115: 1398– 1407, 2007
    1. Li RK, Mickle DA, Weisel RD, Zhang J, Mohabeer MK: In vivo survival and function of transplanted rat cardiomyocytes. Circ Res 78: 283– 288, 1996
    1. Song W, Lu X, Feng Q: Tumor necrosis factor-alpha induces apoptosis via inducible nitric oxide synthase in neonatal mouse cardiomyocytes. Cardiovasc Res 45: 595– 602, 2000
    1. Cook SA, Sugden PH, Clerk A: Regulation of bcl-2 family proteins during development and in response to oxidative stress in cardiac myocytes: association with changes in mitochondrial membrane potential. Circ Res 85: 940– 949, 1999
    1. Noyan-Ashraf MH, Wu L, Wang R, Juurlink BH: Dietary approaches to positively influence fetal determinants of adult health. FASEB J 20: 371– 373, 2006
    1. Tong H, Imahashi K, Steenbergen C, Murphy E: Phosphorylation of glycogen synthase kinase-3beta during preconditioning through a phosphatidylinositol-3-kinase–dependent pathway is cardioprotective. Circ Res 90: 377– 379, 2002
    1. Zhou YQ, Foster FS, Nieman BJ, Davidson L, Chen XJ, Henkelman RM: Comprehensive transthoracic cardiac imaging in mice using ultrasound biomicroscopy with anatomical confirmation by magnetic resonance imaging. Physiol Genomics 18: 232– 244, 2004
    1. Zhou YQ, Zhu Y, Bishop J, Davidson L, Henkelman RM, Bruneau BG, Foster FS: Abnormal cardiac inflow patterns during postnatal development in a mouse model of Holt-Oram syndrome. Am J Physiol Heart Circ Physiol 289: H992– H1001, 2005
    1. Yang Y, Ma Y, Han W, Li J, Xiang Y, Liu F, Ma X, Zhang J, Fu Z, Su YD, Du XJ, Gao XM: Age-related differences in postinfarct left ventricular rupture and remodeling. Am J Physiol Heart Circ Physiol 294: H1815– 1822, 2008
    1. Heymans S, Luttun A, Nuyens D, Theilmeier G, Creemers E, Moons L, Dyspersin GD, Cleutjens JP, Shipley M, Angellilo A, Levi M, Nube O, Baker A, Keshet E, Lupu F, Herbert JM, Smits JF, Shapiro SD, Baes M, Borgers M, Collen D, Daemen MJ, Carmeliet P: Inhibition of plasminogen activators or matrix metalloproteinases prevents cardiac rupture but impairs therapeutic angiogenesis and causes cardiac failure. Nat Med 5: 1135– 1142, 1999
    1. Askari AT, Brennan ML, Zhou X, Drinko J, Morehead A, Thomas JD, Topol EJ, Hazen SL, Penn MS: Myeloperoxidase and plasminogen activator inhibitor 1 play a central role in ventricular remodeling after myocardial infarction. J Exp Med 197: 615– 624, 2003
    1. Yet SF, Tian R, Layne MD, Wang ZY, Maemura K, Solovyeva M, Ith B, Melo LG, Zhang L, Ingwall JS, Dzau VJ, Lee ME, Perrella MA: Cardiac-specific expression of heme oxygenase-1 protects against ischemia and reperfusion injury in transgenic mice. Circ Res 89: 168– 173, 2001
    1. Shiraishi I, Melendez J, Ahn Y, Skavdahl M, Murphy E, Welch S, Schaefer E, Walsh K, Rosenzweig A, Torella D, Nurzynska D, Kajstura J, Leri A, Anversa P, Sussman MA: Nuclear targeting of Akt enhances kinase activity and survival of cardiomyocytes. Circ Res 94: 884– 891, 2004
    1. Heineke J, Molkentin JD: Regulation of cardiac hypertrophy by intracellular signalling pathways. Nat Rev Mol Cell Biol 7: 589– 600, 2006
    1. Juhaszova M, Zorov DB, Kim SH, Pepe S, Fu Q, Fishbein KW, Ziman BD, Wang S, Ytrehus K, Antos CL, Olson EN, Sollott SJ: Glycogen synthase kinase-3beta mediates convergence of protection signaling to inhibit the mitochondrial permeability transition pore. J Clin Invest 113: 1535– 1549, 2004
    1. Liu X, Pachori AS, Ward CA, Davis JP, Gnecchi M, Kong D, Zhang L, Murduck J, Yet SF, Perrella MA, Pratt RE, Dzau VJ, Melo LG: Heme oxygenase-1 (HO-1) inhibits postmyocardial infarct remodeling and restores ventricular function. FASEB J 20: 207– 216, 2006
    1. Burkart EM, Sambandam N, Han X, Gross RW, Courtois M, Gierasch CM, Shoghi K, Welch MJ, Kelly DP: Nuclear receptors PPARbeta/delta and PPARalpha direct distinct metabolic regulatory programs in the mouse heart. J Clin Invest 117: 3930– 3939, 2007
    1. During MJ, Cao L, Zuzga DS, Francis JS, Fitzsimons HL, Jiao X, Bland RJ, Klugmann M, Banks WA, Drucker DJ, Haile CN: Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat Med 9: 1173– 1179, 2003
    1. Li Y, Hansotia T, Yusta B, Ris F, Halban PA, Drucker DJ: Glucagon-like peptide-1 receptor signaling modulates beta cell apoptosis. J Biol Chem 278: 471– 478, 2003
    1. Nikolaidis LA, Elahi D, Shen YT, Shannon RP: Active metabolite of GLP-1 mediates myocardial glucose uptake and improves left ventricular performance in conscious dogs with dilated cardiomyopathy. Am J Physiol Heart Circ Physiol 289: H2401– 2408, 2005
    1. Kavianipour M, Ehlers MR, Malmberg K, Ronquist G, Ryden L, Wikstrom G, Gutniak M: Glucagon-like peptide-1 (7–36) amide prevents the accumulation of pyruvate and lactate in the ischemic and non-ischemic porcine myocardium. Peptides 24: 569– 578, 2003
    1. Inzucchi SE, McGuire DK: New drugs for the treatment of diabetes: part II: incretin-based therapy and beyond. Circulation 117: 574– 584, 2008
    1. Aulinger B, D'Alessio D: Glucagon-like peptide 1: continued advances, new targets and expanding promise as a model therapeutic. Curr Opin Endocrinol Diabetes Obes 14: 68– 73, 2007

Source: PubMed

3
Subscribe