Skip to main content
Top
Published in: Cardiovascular Diabetology 1/2017

Open Access 01-12-2017 | Original investigation

The role of endothelial nitric oxide in the anti-restenotic effects of liraglutide in a mouse model of restenosis

Authors: Hideki Kushima, Yusaku Mori, Masakazu Koshibu, Munenori Hiromura, Kyoko Kohashi, Michishige Terasaki, Tomoyasu Fukui, Tsutomu Hirano

Published in: Cardiovascular Diabetology | Issue 1/2017

Login to get access

Abstract

Background

Previous animal studies have shown that glucagon-like peptide-1 receptor agonists (GLP-1RAs) suppress arterial restenosis, a major complication of angioplasty, presumably through their direct action on vascular smooth muscle cells. However, the contribution of vascular endothelial cells (VECs) to this process remains unknown. In addition, the potential interference caused by severe hyperglycemia and optimal treatment regimen remain to be determined.

Methods

Nine-week-old male C57BL6 (wild-type) and diabetic db/db mice were randomly divided into vehicle or liraglutide treatment groups (Day 1), and subject to femoral artery wire injuries (Day 3). The injured arteries were collected on Day 29 for morphometric analysis. Human umbilical vein endothelial cells (HUVECs) were used for in vitro experiments. One-way ANOVA, followed by Tukey’s test, was used for comparisons.

Results

In wild-type mice, liraglutide treatment (5.7, 17, or 107 nmol/kg/day) dose-dependently reduced the neointimal area (20, 50, and 65%) without inducing systemic effects, and caused an associated decrease in the percentage of vascular proliferating cells. However, these effects were completely abolished by the nitric oxide synthase (NOS) inhibitor N-omega-nitro-l-arginine methyl ester. Next, we investigated the optimal treatment regimen. Early treatment (Days 1–14) was as effective in reducing the neointimal area and vascular cell proliferation as full treatment (Days 1–29), whereas delayed treatment (Days 15–29) was ineffective. In HUVECs, liraglutide treatment dose-dependently stimulated NO production, which was dependent on GLP-1R, cAMP, cAMP-dependent protein kinase, AMP-activated protein kinase (AMPK), and NOS. Subsequently, we investigated the role of liver kinase B (LKB)-1 in this process. Liraglutide increased the phosphorylation of LKB-1, and siRNA-induced LKB-1 knockdown abolished liraglutide-stimulated NO production. In severe hyperglycemic db/db mice, liraglutide treatment also suppressed neointimal hyperplasia, which was accompanied by reductions in vascular cell proliferation and density. Furthermore, liraglutide treatment suppressed hyperglycemia-enhanced vascular inflammation 7 days after arterial injury.

Conclusions

We demonstrate that endothelial cells are targets of liraglutide, and suppress restenosis via endothelial NO. Furthermore, the protective effects are maintained in severe hyperglycemia. Our findings provide an evidence base for a future clinical trial to determine whether treatment with GLP-1RAs represents potentially effective pharmacological therapy following angioplasty in patients with diabetes.
Appendix
Available only for authorised users
Literature
1.
go back to reference Htike ZZ, Zaccardi F, Papamargaritis D, Webb DR, Khunti K, Davies MJ. Efficacy and safety of glucagon-like peptide-1 receptor agonists in type 2 diabetes: a systematic review and mixed-treatment comparison analysis. Diabetes Obes Metab. 2017;19:524–36.CrossRefPubMed Htike ZZ, Zaccardi F, Papamargaritis D, Webb DR, Khunti K, Davies MJ. Efficacy and safety of glucagon-like peptide-1 receptor agonists in type 2 diabetes: a systematic review and mixed-treatment comparison analysis. Diabetes Obes Metab. 2017;19:524–36.CrossRefPubMed
3.
go back to reference Tashiro Y, Sato K, Watanabe T, Nohtomi K, Terasaki M, Nagashima M, et al. A glucagon-like peptide-1 analog liraglutide suppresses macrophage foam cell formation and atherosclerosis. Peptides. 2014;54:19–26.CrossRefPubMed Tashiro Y, Sato K, Watanabe T, Nohtomi K, Terasaki M, Nagashima M, et al. A glucagon-like peptide-1 analog liraglutide suppresses macrophage foam cell formation and atherosclerosis. Peptides. 2014;54:19–26.CrossRefPubMed
4.
go back to reference Nagashima M, Watanabe T, Terasaki M, Tomoyasu M, Nohtomi K, Kim-Kaneyama J, et al. Native incretins prevent the development of atherosclerotic lesions in apolipoprotein E knockout mice. Diabetologia. 2011;54:2649–59.CrossRefPubMedPubMedCentral Nagashima M, Watanabe T, Terasaki M, Tomoyasu M, Nohtomi K, Kim-Kaneyama J, et al. Native incretins prevent the development of atherosclerotic lesions in apolipoprotein E knockout mice. Diabetologia. 2011;54:2649–59.CrossRefPubMedPubMedCentral
5.
go back to reference Bisgaard LS, Bosteen MH, Fink LN, Sørensen CM, Rosendahl A, Mogensen CK, et al. Liraglutide reduces both atherosclerosis and kidney inflammation in moderately uremic LDLr−/− mice. PLoS ONE. 2016. doi:10.1371/journal.pone.0168396. Bisgaard LS, Bosteen MH, Fink LN, Sørensen CM, Rosendahl A, Mogensen CK, et al. Liraglutide reduces both atherosclerosis and kidney inflammation in moderately uremic LDLr−/− mice. PLoS ONE. 2016. doi:10.​1371/​journal.​pone.​0168396.
6.
go back to reference Arakawa M, Mita T, Azuma K, Ebato C, Goto H, Nomiyama T, et al. Inhibition of monocyte adhesion to endothelial cells and attenuation of atherosclerotic lesion by a glucagon-like peptide-1 receptor agonist, exendin-4. Diabetes. 2010;59:1030–107.CrossRefPubMedPubMedCentral Arakawa M, Mita T, Azuma K, Ebato C, Goto H, Nomiyama T, et al. Inhibition of monocyte adhesion to endothelial cells and attenuation of atherosclerotic lesion by a glucagon-like peptide-1 receptor agonist, exendin-4. Diabetes. 2010;59:1030–107.CrossRefPubMedPubMedCentral
7.
go back to reference Jojima T, Uchida K, Akimoto K, Tomotsune T, Yanagi K, Iijima T, et al. Liraglutide, a GLP-1 receptor agonist, inhibits vascular smooth muscle cell proliferation by enhancing AMP-activated protein kinase and cell cycle regulation, and delays atherosclerosis in ApoE deficient mice. Atherosclerosis. 2017;261:44–51.CrossRefPubMed Jojima T, Uchida K, Akimoto K, Tomotsune T, Yanagi K, Iijima T, et al. Liraglutide, a GLP-1 receptor agonist, inhibits vascular smooth muscle cell proliferation by enhancing AMP-activated protein kinase and cell cycle regulation, and delays atherosclerosis in ApoE deficient mice. Atherosclerosis. 2017;261:44–51.CrossRefPubMed
8.
go back to reference Gaspari T, Welungoda I, Widdop RE, Simpson RW, Dear AE. The GLP-1 receptor agonist liraglutide inhibits progression of vascular disease via effects on atherogenesis, plaque stability and endothelial function in an ApoE(−/−) mouse model. Diab Vasc Dis Res. 2013;10:353–60.CrossRefPubMed Gaspari T, Welungoda I, Widdop RE, Simpson RW, Dear AE. The GLP-1 receptor agonist liraglutide inhibits progression of vascular disease via effects on atherogenesis, plaque stability and endothelial function in an ApoE(−/−) mouse model. Diab Vasc Dis Res. 2013;10:353–60.CrossRefPubMed
9.
go back to reference Wang Y, Parlevliet ET, Geerling JJ, van der Tuin SJ, Zhang H, Bieghs V, et al. Exendin-4 decreases liver inflammation and atherosclerosis development simultaneously by reducing macrophage infiltration. Br J Pharmacol. 2014;171:723–34.CrossRefPubMedPubMedCentral Wang Y, Parlevliet ET, Geerling JJ, van der Tuin SJ, Zhang H, Bieghs V, et al. Exendin-4 decreases liver inflammation and atherosclerosis development simultaneously by reducing macrophage infiltration. Br J Pharmacol. 2014;171:723–34.CrossRefPubMedPubMedCentral
10.
go back to reference Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375:311–22.CrossRefPubMedPubMedCentral Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375:311–22.CrossRefPubMedPubMedCentral
11.
go back to reference Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2016;375:1834–44.CrossRefPubMed Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2016;375:1834–44.CrossRefPubMed
12.
go back to reference Goto H, Nomiyama T, Mita T, Yasunari E, Azuma K, Komiya K, et al. Exendin-4, a glucagon-like peptide-1 receptor agonist, reduces intimal thickening after vascular injury. Biochem Biophys Res Commun. 2011;405:79–84.CrossRefPubMed Goto H, Nomiyama T, Mita T, Yasunari E, Azuma K, Komiya K, et al. Exendin-4, a glucagon-like peptide-1 receptor agonist, reduces intimal thickening after vascular injury. Biochem Biophys Res Commun. 2011;405:79–84.CrossRefPubMed
13.
go back to reference Hirata Y, Kurobe H, Nishio C, Tanaka K, Fukuda D, Uematsu E, et al. Exendin-4, a glucagon-like peptide-1 receptor agonist, attenuates neointimal hyperplasia after vascular injury. Eur J Pharmacol. 2013;699:106–11.CrossRefPubMed Hirata Y, Kurobe H, Nishio C, Tanaka K, Fukuda D, Uematsu E, et al. Exendin-4, a glucagon-like peptide-1 receptor agonist, attenuates neointimal hyperplasia after vascular injury. Eur J Pharmacol. 2013;699:106–11.CrossRefPubMed
14.
go back to reference Eriksson L, Saxelin R, Röhl S, Roy J, Caidahl K, Nyström T, et al. Glucagon-like peptide-1 receptor activation does not affect re-endothelialization but reduces intimal hyperplasia via direct effects on smooth muscle cells in a nondiabetic model of arterial injury. J Vasc Res. 2015;52:41–52.CrossRefPubMed Eriksson L, Saxelin R, Röhl S, Roy J, Caidahl K, Nyström T, et al. Glucagon-like peptide-1 receptor activation does not affect re-endothelialization but reduces intimal hyperplasia via direct effects on smooth muscle cells in a nondiabetic model of arterial injury. J Vasc Res. 2015;52:41–52.CrossRefPubMed
15.
go back to reference Lim S, Lee GY, Park HS, Lee DH, Tae Jung O, Kyoung Min K, et al. Attenuation of carotid neointimal formation after direct delivery of a recombinant adenovirus expressing glucagon-like peptide-1 in diabetic rats. Cardiovasc Res. 2017;113:183–94.CrossRefPubMed Lim S, Lee GY, Park HS, Lee DH, Tae Jung O, Kyoung Min K, et al. Attenuation of carotid neointimal formation after direct delivery of a recombinant adenovirus expressing glucagon-like peptide-1 in diabetic rats. Cardiovasc Res. 2017;113:183–94.CrossRefPubMed
16.
go back to reference Korshunov VA, Schwartz SM, Berk BC. Vascular remodeling: hemodynamic and biochemical mechanisms underlying Glagov’s phenomenon. Arterioscler Thromb Vasc Biol. 2007;27:1722–8.CrossRefPubMed Korshunov VA, Schwartz SM, Berk BC. Vascular remodeling: hemodynamic and biochemical mechanisms underlying Glagov’s phenomenon. Arterioscler Thromb Vasc Biol. 2007;27:1722–8.CrossRefPubMed
17.
go back to reference Kokkinidis DG, Waldo SW, Armstrong EJ. Treatment of coronary artery in-stent restenosis. Expert Rev Cardiovasc Ther. 2017;15:191–202.CrossRefPubMed Kokkinidis DG, Waldo SW, Armstrong EJ. Treatment of coronary artery in-stent restenosis. Expert Rev Cardiovasc Ther. 2017;15:191–202.CrossRefPubMed
18.
go back to reference Kang SH, Park KW, Kang DY, Lim WH, Park KT, Han JK, et al. Biodegradable-polymer, drug-eluting stents vs. bare metal stents vs. durable-polymer drug-eluting stents: a systematic review and Bayesian approach network meta-analysis. Eur Heart J. 2014;35:1147–58.CrossRefPubMed Kang SH, Park KW, Kang DY, Lim WH, Park KT, Han JK, et al. Biodegradable-polymer, drug-eluting stents vs. bare metal stents vs. durable-polymer drug-eluting stents: a systematic review and Bayesian approach network meta-analysis. Eur Heart J. 2014;35:1147–58.CrossRefPubMed
19.
go back to reference Aronson D, Edelman ER. Revascularization for coronary artery disease in diabetes mellitus: angioplasty, stents and coronary artery bypass grafting. Rev Endocr Metab Disord. 2010;11:75–86.CrossRefPubMedPubMedCentral Aronson D, Edelman ER. Revascularization for coronary artery disease in diabetes mellitus: angioplasty, stents and coronary artery bypass grafting. Rev Endocr Metab Disord. 2010;11:75–86.CrossRefPubMedPubMedCentral
20.
go back to reference Stettler C, Allemann S, Wandel S, Kastrati A, Morice MC, Schomig A, et al. Drug eluting and bare metal stents in people with and without diabetes: collaborative network meta-analysis. BMJ. 2008;337:a1331–4.CrossRefPubMedPubMedCentral Stettler C, Allemann S, Wandel S, Kastrati A, Morice MC, Schomig A, et al. Drug eluting and bare metal stents in people with and without diabetes: collaborative network meta-analysis. BMJ. 2008;337:a1331–4.CrossRefPubMedPubMedCentral
21.
go back to reference Sata M, Maejima Y, Adachi F, Fukino K, Saiura A, Sugiura S, et al. A mouse model of vascular injury that induces rapid onset of medial cell apoptosis followed by reproducible neointimal hyperplasia. J Mol Cell Cardiol. 2000;32:2097–104.CrossRefPubMed Sata M, Maejima Y, Adachi F, Fukino K, Saiura A, Sugiura S, et al. A mouse model of vascular injury that induces rapid onset of medial cell apoptosis followed by reproducible neointimal hyperplasia. J Mol Cell Cardiol. 2000;32:2097–104.CrossRefPubMed
22.
go back to reference Shi L, Ji Y, Jiang X, Zhou L, Xu Y, Li Y, et al. Liraglutide attenuates high glucose-induced abnormal cell migration, proliferation, and apoptosis of vascular smooth muscle cells by activating the GLP-1 receptor, and inhibiting ERK1/2 and PI3 K/Akt signaling pathways. Cardiovasc Diabetol. 2015;14:18.CrossRefPubMedPubMedCentral Shi L, Ji Y, Jiang X, Zhou L, Xu Y, Li Y, et al. Liraglutide attenuates high glucose-induced abnormal cell migration, proliferation, and apoptosis of vascular smooth muscle cells by activating the GLP-1 receptor, and inhibiting ERK1/2 and PI3 K/Akt signaling pathways. Cardiovasc Diabetol. 2015;14:18.CrossRefPubMedPubMedCentral
24.
go back to reference Otsuka F, Finn AV, Yazdani SK, Nakano M, Kolodgie FD, Virmani R. The importance of the endothelium in atherothrombosis and coronary stenting. Nat Rev Cardiol. 2012;9:439–53.CrossRefPubMed Otsuka F, Finn AV, Yazdani SK, Nakano M, Kolodgie FD, Virmani R. The importance of the endothelium in atherothrombosis and coronary stenting. Nat Rev Cardiol. 2012;9:439–53.CrossRefPubMed
25.
go back to reference Farooq V, Gogas BD, Serruys PW. Restenosis: delineating the numerous causes of drug-eluting stent restenosis. Circ Cardiovasc Interv. 2011;4:195–205.CrossRefPubMed Farooq V, Gogas BD, Serruys PW. Restenosis: delineating the numerous causes of drug-eluting stent restenosis. Circ Cardiovasc Interv. 2011;4:195–205.CrossRefPubMed
26.
go back to reference Kauser K, da Cunha V, Fitch R, Mallari C, Rubanyi GM. Role of endogenous nitric oxide in progression of atherosclerosis in apolipoprotein E-deficient mice. Am J Physiol Heart Circ Physiol. 2000;278:H1679–85.PubMed Kauser K, da Cunha V, Fitch R, Mallari C, Rubanyi GM. Role of endogenous nitric oxide in progression of atherosclerosis in apolipoprotein E-deficient mice. Am J Physiol Heart Circ Physiol. 2000;278:H1679–85.PubMed
27.
go back to reference Ke J, Liu Y, Yang J, Lu R, Tian Q, Hou W, et al. Synergistic effects of metformin with liraglutide against endothelial dysfunction through GLP-1 receptor and PKA signalling pathway. Sci Rep. 2017. doi:10.1038/srep41085. Ke J, Liu Y, Yang J, Lu R, Tian Q, Hou W, et al. Synergistic effects of metformin with liraglutide against endothelial dysfunction through GLP-1 receptor and PKA signalling pathway. Sci Rep. 2017. doi:10.​1038/​srep41085.
28.
go back to reference Ding L, Zhang J. Glucagon-like peptide-1 activates endothelial nitric oxide synthase in human umbilical vein endothelial cells. Acta Pharmacol Sin. 2012;33:75–81.CrossRefPubMed Ding L, Zhang J. Glucagon-like peptide-1 activates endothelial nitric oxide synthase in human umbilical vein endothelial cells. Acta Pharmacol Sin. 2012;33:75–81.CrossRefPubMed
29.
go back to reference Koska J, Sands M, Burciu C, D’Souza KM, Raravikar K, Liu J, et al. Exenatide protects against glucose- and lipid-induced endothelial dysfunction: evidence for direct vasodilation effect of GLP-1 receptor agonists in humans. Diabetes. 2015;64:2624–35.CrossRefPubMedPubMedCentral Koska J, Sands M, Burciu C, D’Souza KM, Raravikar K, Liu J, et al. Exenatide protects against glucose- and lipid-induced endothelial dysfunction: evidence for direct vasodilation effect of GLP-1 receptor agonists in humans. Diabetes. 2015;64:2624–35.CrossRefPubMedPubMedCentral
30.
go back to reference Liu L, Liu J, Wong WT, Tian XY, Lau CW, Wang YX, et al. Dipeptidyl peptidase 4 inhibitor sitagliptin protects endothelial function in hypertension through a glucagon-like peptide 1-dependent mechanism. Hypertension. 2012;60:833–41.CrossRefPubMed Liu L, Liu J, Wong WT, Tian XY, Lau CW, Wang YX, et al. Dipeptidyl peptidase 4 inhibitor sitagliptin protects endothelial function in hypertension through a glucagon-like peptide 1-dependent mechanism. Hypertension. 2012;60:833–41.CrossRefPubMed
31.
go back to reference Kjems LL, Holst JJ, Vølund A, Madsbad S. The influence of GLP-1 on glucose-stimulated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic subjects. Diabetes. 2003;52:380–6.CrossRefPubMed Kjems LL, Holst JJ, Vølund A, Madsbad S. The influence of GLP-1 on glucose-stimulated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic subjects. Diabetes. 2003;52:380–6.CrossRefPubMed
32.
go back to reference Kohashi K, Hiromura M, Mori Y, Terasaki M, Watanabe T, Kushima H, et al. A dipeptidyl peptidase-4 inhibitor but not incretins suppresses abdominal aortic aneurysms in angiotensin II-infused apolipoprotein E-null mice. J Atheroscler Thromb. 2016;23:441–54.CrossRefPubMed Kohashi K, Hiromura M, Mori Y, Terasaki M, Watanabe T, Kushima H, et al. A dipeptidyl peptidase-4 inhibitor but not incretins suppresses abdominal aortic aneurysms in angiotensin II-infused apolipoprotein E-null mice. J Atheroscler Thromb. 2016;23:441–54.CrossRefPubMed
33.
go back to reference Igawa S, Hayashi I, Tanaka N, Hiruma H, Majima M, Kawakami T, et al. Nitric oxide generated by iNOS reduces deformability of lewis lung carcinoma cells. Cancer Sci. 2004;95:342–7.CrossRefPubMed Igawa S, Hayashi I, Tanaka N, Hiruma H, Majima M, Kawakami T, et al. Nitric oxide generated by iNOS reduces deformability of lewis lung carcinoma cells. Cancer Sci. 2004;95:342–7.CrossRefPubMed
34.
go back to reference Yu X, Li F, Klussmann E, Stallone JN, Han G. G protein-coupled estrogen receptor 1 mediates relaxation of coronary arteries via cAMP/PKA-dependent activation of MLCP. Am J Physiol Endocrinol Metab. 2014;15(307):E398–407.CrossRef Yu X, Li F, Klussmann E, Stallone JN, Han G. G protein-coupled estrogen receptor 1 mediates relaxation of coronary arteries via cAMP/PKA-dependent activation of MLCP. Am J Physiol Endocrinol Metab. 2014;15(307):E398–407.CrossRef
35.
go back to reference Spirli C, Okolicsanyi S, Fiorotto R, Fabris L, Cadamuro M, Lecchi S, et al. ERK1/2-dependent vascular endothelial growth factor signaling sustains cyst growth in polycystin-2 defective mice. Gastroenterology. 2010;138:360–71.CrossRefPubMed Spirli C, Okolicsanyi S, Fiorotto R, Fabris L, Cadamuro M, Lecchi S, et al. ERK1/2-dependent vascular endothelial growth factor signaling sustains cyst growth in polycystin-2 defective mice. Gastroenterology. 2010;138:360–71.CrossRefPubMed
36.
go back to reference Almahariq M, Tsalkova T, Mei FC, Chen H, Zhou J, Sastry SK, et al. A novel EPAC-specific inhibitor suppresses pancreatic cancer cell migration and invasion. Mol Pharmacol. 2013;83:122–8.CrossRefPubMedPubMedCentral Almahariq M, Tsalkova T, Mei FC, Chen H, Zhou J, Sastry SK, et al. A novel EPAC-specific inhibitor suppresses pancreatic cancer cell migration and invasion. Mol Pharmacol. 2013;83:122–8.CrossRefPubMedPubMedCentral
37.
go back to reference Calay D, Vind-Kezunovic D, Frankart A, Lambert S, Poumay Y, Gniadecki R. Inhibition of Akt signaling by exclusion from lipid rafts in normal and transformed epidermal keratinocytes. J Invest Dermatol. 2010;130:1136–45.CrossRefPubMed Calay D, Vind-Kezunovic D, Frankart A, Lambert S, Poumay Y, Gniadecki R. Inhibition of Akt signaling by exclusion from lipid rafts in normal and transformed epidermal keratinocytes. J Invest Dermatol. 2010;130:1136–45.CrossRefPubMed
38.
go back to reference Wang W, Diamond SL. Does elevated nitric oxide production enhance the release of prostacyclin from shear stressed aortic endothelial cells? Biochem Biophys Res Commun. 1997;233:748–51.CrossRefPubMed Wang W, Diamond SL. Does elevated nitric oxide production enhance the release of prostacyclin from shear stressed aortic endothelial cells? Biochem Biophys Res Commun. 1997;233:748–51.CrossRefPubMed
39.
go back to reference Hawley SA, Pan DA, Mustard KJ, Ross L, Bain J, Edelman AM, et al. Calmodulin-dependent protein kinase kinase-beta is an alternative upstream kinase for AMP-activated protein kinase. Cell Metab. 2005;2:9–19.CrossRefPubMed Hawley SA, Pan DA, Mustard KJ, Ross L, Bain J, Edelman AM, et al. Calmodulin-dependent protein kinase kinase-beta is an alternative upstream kinase for AMP-activated protein kinase. Cell Metab. 2005;2:9–19.CrossRefPubMed
40.
go back to reference Pereira S, Park E, Mori Y, Haber CA, Han P, Uchida T, et al. FFA-induced hepatic insulin resistance in vivo is mediated by PKCδ, NADPH oxidase, and oxidative stress. Am J Physiol Endocrinol Metab. 2014;307:E34–46.CrossRefPubMedPubMedCentral Pereira S, Park E, Mori Y, Haber CA, Han P, Uchida T, et al. FFA-induced hepatic insulin resistance in vivo is mediated by PKCδ, NADPH oxidase, and oxidative stress. Am J Physiol Endocrinol Metab. 2014;307:E34–46.CrossRefPubMedPubMedCentral
41.
go back to reference Peotta VA, Vasquez EC, Meyrelles SS. Cardiovascular neural reflexes in l-NAME-induced hypertension in mice. Hypertension. 2001;38:555–9.CrossRefPubMed Peotta VA, Vasquez EC, Meyrelles SS. Cardiovascular neural reflexes in l-NAME-induced hypertension in mice. Hypertension. 2001;38:555–9.CrossRefPubMed
42.
go back to reference Guo J, Breen DM, Pereira TJ, Dalvi PS, Zhang H, Mori Y, et al. The effect of insulin to decrease neointimal growth after arterial injury is endothelial nitric oxide synthase-dependent. Atherosclerosis. 2015;241:111–20.CrossRefPubMed Guo J, Breen DM, Pereira TJ, Dalvi PS, Zhang H, Mori Y, et al. The effect of insulin to decrease neointimal growth after arterial injury is endothelial nitric oxide synthase-dependent. Atherosclerosis. 2015;241:111–20.CrossRefPubMed
43.
go back to reference Pearce CG, Najjar SF, Kapadia MR, Murar J, Eng J, Lyle B, et al. Beneficial effect of a short-acting NO donor for the prevention of neointimal hyperplasia. Free Radic Biol Med. 2008;44:73–81.CrossRefPubMed Pearce CG, Najjar SF, Kapadia MR, Murar J, Eng J, Lyle B, et al. Beneficial effect of a short-acting NO donor for the prevention of neointimal hyperplasia. Free Radic Biol Med. 2008;44:73–81.CrossRefPubMed
44.
go back to reference Chen L, Daum G, Forough R, Clowes M, Walter U, Clowes AW. Overexpression of human endothelial nitric oxide synthase in rat vascular smooth muscle cells and in balloon-injured carotid artery. Circ Res. 1998;82:862–70.CrossRefPubMed Chen L, Daum G, Forough R, Clowes M, Walter U, Clowes AW. Overexpression of human endothelial nitric oxide synthase in rat vascular smooth muscle cells and in balloon-injured carotid artery. Circ Res. 1998;82:862–70.CrossRefPubMed
45.
go back to reference Xu G, Kaneto H, Laybutt DR, Duvivier-Kali VF, Trivedi N, Suzuma K, et al. Downregulation of GLP-1 and GIP receptor expression by hyperglycemia: possible contribution to impaired incretin effects in diabetes. Diabetes. 2007;56:1551–8.CrossRefPubMed Xu G, Kaneto H, Laybutt DR, Duvivier-Kali VF, Trivedi N, Suzuma K, et al. Downregulation of GLP-1 and GIP receptor expression by hyperglycemia: possible contribution to impaired incretin effects in diabetes. Diabetes. 2007;56:1551–8.CrossRefPubMed
46.
go back to reference Panjwani N, Mulvihill EE, Longuet C, Yusta B, Campbell JE, Brown TJ, et al. GLP-1 receptor activation indirectly reduces hepatic lipid accumulation but does not attenuate development of atherosclerosis in diabetic male ApoE(−/−) mice. Endocrinology. 2013;154:127–39.CrossRefPubMed Panjwani N, Mulvihill EE, Longuet C, Yusta B, Campbell JE, Brown TJ, et al. GLP-1 receptor activation indirectly reduces hepatic lipid accumulation but does not attenuate development of atherosclerosis in diabetic male ApoE(−/−) mice. Endocrinology. 2013;154:127–39.CrossRefPubMed
47.
go back to reference Libby P, Schwartz D, Brogi E, Tanaka H, Clinton SK. A cascade model for restenosis. A special case of atherosclerosis progression. Circulation. 1992;86:III47–52.CrossRefPubMed Libby P, Schwartz D, Brogi E, Tanaka H, Clinton SK. A cascade model for restenosis. A special case of atherosclerosis progression. Circulation. 1992;86:III47–52.CrossRefPubMed
48.
go back to reference Khan R, Agrotis A, Bobik A. Understanding the role of transforming growth factor-beta1 in intimal thickening after vascular injury. Cardiovasc Res. 2007;74:223–34.CrossRefPubMed Khan R, Agrotis A, Bobik A. Understanding the role of transforming growth factor-beta1 in intimal thickening after vascular injury. Cardiovasc Res. 2007;74:223–34.CrossRefPubMed
49.
50.
go back to reference Förstermann U, Sessa WC. Nitric oxide synthases: regulation and function. Eur Heart J. 2012;33:829–37.CrossRefPubMed Förstermann U, Sessa WC. Nitric oxide synthases: regulation and function. Eur Heart J. 2012;33:829–37.CrossRefPubMed
51.
go back to reference Tan X, Feng L, Huang X, Yang Y, Yang C, Gao Y. Histone deacetylase inhibitors promote eNOS expression in vascular smooth muscle cells and suppress hypoxia-induced cell growth. J Cell Mol Med. 2017. doi:10.1111/jcmm.13122. Tan X, Feng L, Huang X, Yang Y, Yang C, Gao Y. Histone deacetylase inhibitors promote eNOS expression in vascular smooth muscle cells and suppress hypoxia-induced cell growth. J Cell Mol Med. 2017. doi:10.​1111/​jcmm.​13122.
52.
go back to reference Lee Mei-Yueh, Tsai Kun-Bow, Hsu Jong-Hau, Shin Shyi-Jang, Jiunn-Ren Wu, Yeh Jwu-Lai. Liraglutide prevents and reverses monocrotaline-induced pulmonary arterial hypertension by suppressing ET-1 and enhancing eNOS/sGC/PKG pathways. Sci Rep. 2016;6:31788.CrossRefPubMedPubMedCentral Lee Mei-Yueh, Tsai Kun-Bow, Hsu Jong-Hau, Shin Shyi-Jang, Jiunn-Ren Wu, Yeh Jwu-Lai. Liraglutide prevents and reverses monocrotaline-induced pulmonary arterial hypertension by suppressing ET-1 and enhancing eNOS/sGC/PKG pathways. Sci Rep. 2016;6:31788.CrossRefPubMedPubMedCentral
53.
go back to reference Feil R, Wagner J, Metzger D, Chambon P. Regulation of Cre recombinase activity by mutated estrogen receptor ligand-binding domains. Biochem Biophys Res Commun. 1997;237:752–7.CrossRefPubMed Feil R, Wagner J, Metzger D, Chambon P. Regulation of Cre recombinase activity by mutated estrogen receptor ligand-binding domains. Biochem Biophys Res Commun. 1997;237:752–7.CrossRefPubMed
54.
go back to reference Gossen M, Bujard H. Tight control of gene expression in mammalian cells by tetracycline-responsive promoters. Proc Natl Acad Sci USA. 1992;89:5547–51.CrossRefPubMedPubMedCentral Gossen M, Bujard H. Tight control of gene expression in mammalian cells by tetracycline-responsive promoters. Proc Natl Acad Sci USA. 1992;89:5547–51.CrossRefPubMedPubMedCentral
55.
go back to reference Zhou X, Huang CH, Lao J, Pocai A, Forrest G, Price O, et al. Acute hemodynamic and renal effects of glucagon-like peptide 1 analog and dipeptidyl peptidase-4 inhibitor in rats. Cardiovasc Diabetol. 2015;14:29.CrossRefPubMedPubMedCentral Zhou X, Huang CH, Lao J, Pocai A, Forrest G, Price O, et al. Acute hemodynamic and renal effects of glucagon-like peptide 1 analog and dipeptidyl peptidase-4 inhibitor in rats. Cardiovasc Diabetol. 2015;14:29.CrossRefPubMedPubMedCentral
56.
go back to reference Nielsen R, Jorsal A, Iversen P, Tolbod LP, Bouchelouche K, Sørensen J, et al. Effect of liraglutide on myocardial glucose uptake and blood flow in stable chronic heart failure patients: a double-blind, randomized, placebo-controlled LIVE sub-study. J Nucl Cardiol. 2017. doi:10.1007/s12350-017-1000-2 (Epub ahead of print).PubMed Nielsen R, Jorsal A, Iversen P, Tolbod LP, Bouchelouche K, Sørensen J, et al. Effect of liraglutide on myocardial glucose uptake and blood flow in stable chronic heart failure patients: a double-blind, randomized, placebo-controlled LIVE sub-study. J Nucl Cardiol. 2017. doi:10.​1007/​s12350-017-1000-2 (Epub ahead of print).PubMed
57.
go back to reference Pfeffer MA, Claggett B, Diaz R, Dickstein K, Gerstein HC, Køber LV, et al. Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med. 2015;373:2247–57.CrossRefPubMed Pfeffer MA, Claggett B, Diaz R, Dickstein K, Gerstein HC, Køber LV, et al. Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med. 2015;373:2247–57.CrossRefPubMed
58.
go back to reference Kumarathurai P, Anholm C, Nielsen OW, Kristiansen OP, Mølvig J, Madsbad S, et al. Effects of the glucagon-like peptide-1 receptor agonist liraglutide on systolic function in patients with coronary artery disease and type 2 diabetes: a randomized double-blind placebo-controlled crossover study. Cardiovasc Diabetol. 2016;15:105.CrossRefPubMedPubMedCentral Kumarathurai P, Anholm C, Nielsen OW, Kristiansen OP, Mølvig J, Madsbad S, et al. Effects of the glucagon-like peptide-1 receptor agonist liraglutide on systolic function in patients with coronary artery disease and type 2 diabetes: a randomized double-blind placebo-controlled crossover study. Cardiovasc Diabetol. 2016;15:105.CrossRefPubMedPubMedCentral
Metadata
Title
The role of endothelial nitric oxide in the anti-restenotic effects of liraglutide in a mouse model of restenosis
Authors
Hideki Kushima
Yusaku Mori
Masakazu Koshibu
Munenori Hiromura
Kyoko Kohashi
Michishige Terasaki
Tomoyasu Fukui
Tsutomu Hirano
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Cardiovascular Diabetology / Issue 1/2017
Electronic ISSN: 1475-2840
DOI
https://doi.org/10.1186/s12933-017-0603-x

Other articles of this Issue 1/2017

Cardiovascular Diabetology 1/2017 Go to the issue