Liraglutide, a glucagon‑like peptide‑1 analog, inhibits high glucose‑induced oxidative stress and apoptosis in neonatal rat cardiomyocytes

  • Authors:
    • Lihui Zhang
    • Caige Li
    • Qiuxiao Zhu
    • Na Li
    • Hong Zhou
  • View Affiliations

  • Published online on: March 13, 2019     https://doi.org/10.3892/etm.2019.7388
  • Pages: 3734-3740
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cardiomyocyte apoptosis serves an important role in diabetic cardiomyopathy. Liraglutide, a glucagon‑like peptide‑1 analog, has been indicated to exert a cardioprotective effect. However, the role of liraglutide on cardiomyocyte apoptosis in hyperglycemia is not fully understood. The aim of the current study was to assess whether liraglutide protects against high glucose (HG)‑induced cardiomyocyte apoptosis in vitro. Sprague‑Dawley neonatal rat cardiomyocytes were cultured in Dulbecco's modified Eagle's medium, supplemented with 5.5 or 25 mmol/l D‑glucose or 5.5 mmol/l D‑glucose + 19.5 mmol/l mannitol, in the presence or absence of liraglutide (10 or 100 nmol/l). Cell viability was assessed via an MTT assay and early apoptosis rates were assessed via flow cytometry. Superoxide dismutase (SOD) activity and malondialdehyde (MDA) content in cell supernatants were measured. Bcl‑2 associated X (Bax), B‑cell lymphoma‑2 (Bcl‑2) and cleaved/full caspase‑3 protein levels were determined via western blotting. The results revealed that liraglutide effectively inhibited the HG‑induced increase in early apoptosis and MDA content and markedly increased SOD activity. Furthermore, liraglutide markedly inhibited the HG‑induced increase in Bax and cleaved caspase‑3 protein expression, and upregulated the expression of Bcl‑2. The present study demonstrated that liraglutide suppressed HG‑induced oxidative stress and cardiomyocyte apoptosis. Thus, the anti‑apoptotic actions of liraglutide may be attributable, in part, to the inhibition of Bax, the inhibition of caspase‑3 activation and the upregualtion of Bcl‑2.

Introduction

Diabetic cardiomyopathy (DCM) is a serious complication of diabetes, which increases the mortality of patients (1). DCM is defined as left ventricular dysfunction that occurs independently of hypertension and coronary artery atherosclerosis and is a cause of heart failure in patients with diabetes (2). Increased oxidative stress and cardiomyocyte apoptosis have been implicated in the development of DCM (3,4). Therefore, inhibiting cardiomyocyte apoptosis is a key step in the prevention of DCM. Glucose-lowering agents that decrease the risk of major cardiovascular events would thus be considered important (5). Glucagon-like peptide-1 (GLP-1) is a 30-amino acid gut hormone that is secreted from intestinal endocrine L cells, which stimulates insulin secretion, inhibits glucagon secretion and inhibits gastric emptying, causing postprandial euglycemia and body weight reduction (6). Multiple GLP-1 analogs have been developed and one such agent, liraglutide, was approved for the treatment of type 2 diabetes and obesity (7). Growing evidence has indicated that GLP-1 analogs have the potential to reduce cardiac inflammation, limit infarct size and mitigate ischemic-reperfusion injury in animals with experimental myocardial infarction (MI) (810). Recently, several cardiovascular studies have documented the reduction of major adverse cardiovascular events and cardiovascular mortality in patients with type 2 diabetes or preexisting cardiovascular disease, via treatment with liraglutide and semaglutide (11,12). The beneficial effects exhibited by liraglutide and semaglutide may be associated with reductions in hemoglobin A1c, body weight, systolic blood pressure and lipoproteins (1113). However, traditional atherogenic risk factor modifications alone cannot explain the overall benefits observed, indicating that additional mechanisms may occur (1113). The favorable effects of liraglutide on oxidative stress and carotid atherosclerosis in patients with diabetes has been previously reported (14,15). Various studies have also revealed that liraglutide exhibits protective myocardial actions in diabetic animal models in vivo (16,17). However, the results of in vivo studies may have been influenced by many factors, including metabolic and environmental factors as well as the anti-atherosclerotic effect of liraglutide (15,17,18). Furthermore, few in vitro reports detail the role of liraglutide on cardiomyocytes in a high glucose (HG) state. Therefore, to assess the possible mechanism of liraglutide on myocardial protection in diabetes, the present study determined the effects of liraglutide on HG-induced oxidative stress and apoptosis in neonatal rat cardiomyocytes in vitro.

Materials and methods

Primary culture of rat myocytes

The animal protocol was reviewed and approved by the Laboratory Animal Ethical and Welfare Committee of Hebei Medical University (Shijiazhuang, China; approval no. IACUC-Hebmu-20160027). A total of 160 Sprague-Dawley (SD) rats (age, 3 days; weight, 8–10 g; 80 males and 80 females) were obtained from the Laboratory Animal Center of Hebei Medical University and used in the experiment immediately. Rat myocytes were prepared as previously described (19). Neonatal SD rats were euthanized using carbon dioxide (CO2) and the flow rate displaced 20% of the chamber volume/minute. Rats were exposed to 50% CO2 until they were euthanized, at which point they were decapitated. Rat ventricles were subsequently removed, minced and digested in PBS (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 0.1% trypsin (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and 0.04% type II collagenase (Invitrogen; Thermo Fisher Scientific, Inc.) eight to 10 times. Samples were centrifuged (320 × g, 37°C, 5 min) and suspended in Dulbecco's modified Eagle's medium (DMEM; Sigma-Aldrich; Merck KGaA) containing 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and 5.5 mmol/l D-glucose. The suspension was maintained in DMEM for 2 h in a humidified atmosphere of 95% air and 5% CO2 at 37°C, which was used to further increase the ratio of rat myocytes to non-cardiomyocytes. Unattached myocytes were plated at 1×106 cells/cm2 in the aforementioned medium supplemented with 0.1 mM bromodeoxyuridine (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C for 48 h. Myocytes were then placed in Lonza 12-725F UltraCULTURE serum-free medium (Lonza Walkersville, Inc., Walkersville, MD, USA) at 37°C for 24 h prior to experimentation. Rat myocytes were confirmed via morphological examination on a light microscope at a magnification of ×400 and staining with an anti-α-sarcomeric actin (α-SMA) antibodies (cat. no. LM-10196R-FITC; dilution, 1:200; Sigma-Aldrich; Merck KGaA) overnight at 4°C. The α-SMA-positive cells were verified to be myocytes, ~95% of cells were identified as rat myocytes.

Drug treatments

Liraglutide, a GLP-1 analog, was obtained from Novo Nordisk Ltd. (Gatwick, UK). Liraglutide at a concentration of 10 and 100 nmol/l was selected according to previous studies (9,18) and cardiomyocyte viability determined in the present study (Fig. 1). When cardiomyocytes reached a confluence of 80%, cells were pre-incubated at 37°C in the presence or absence of 10 or 100 nmol/l liraglutide for 30 min. DMEM was then replaced with DMEM containing 5.5 mmol/l D-glucose [normal glucose (NG) group], 25 mmol/l D-glucose (HG group) or mannitol containing 5.5 mmol/l D-glucose and 19.5 mmol/l mannitol [osmotic control (OSM) group]. Myocytes of the HG group were further incubated at 37°C for 24 h in the presence of liraglutide (10 and 100 nmol/l; named the HG + 10 nM liraglutide and HG + 100 nM liraglutide groups, respectively).

Cell survival assay

Cell viability was assessed via an MTT assay (Sigma-Aldrich; Merck KGaA). Myocytes were plated at 1×104 cells/well in 96-well plates and 20 µl of 5 mg/ml MTT was added to each well and incubated for 4 h at 37°C. Samples were then were solubilized with 150 µl dimethyl sulfoxide. Absorbance was read at 490 nm. Each experiment was repeated three times and three independent experiments were performed.

Flow cytometry

A fluorescein isothiocyanate (FITC) Annexin-V apoptosis detection kit (BD Biosciences, San Jose, CA, USA) was used to detect apoptosis in neonatal rat cardiomyocytes following various treatments. Cardiomyocytes were washed in PBS three times and resuspended in 400 µl of binding buffer with FITC Annexin V and propidium iodide (PI, 5 µl of each). Cell suspensions were incubated for 15 min at room temperature in the dark and analyzed via flow cytometry within 1 h. FlowJo software (version 7.6; FlowJo LLC, Ashland, OR, USA) was used for data acquisition. Positive Annexin V-FITC and negative PI cells were identified as early apoptotic cells. Apoptosis rate was calculated as the number of early apoptotic cells relative to the total number of cells. Each experiment was repeated three times and three independent experiments were performed.

Superoxide dismutase (SOD) activity and malondialdehyde (MDA) content

Following cell treatments, the supernatant was collected to measure SOD activity and MDA content. Measurements were obtained using commercial kits (SOD, cat. no. A001-1-1; MDA, cat. no. A003-1; Nanjing Jiancheng Biological Engineering Institute, Nanjing, China) in accordance with the manufacturer's protocol. Each experiment was repeated three times and three independent experiments were performed.

Western blotting

Myocytes were grown at 1×106 cells/cm2 in culture dishes as aforementioned. Following rinsing with cold D-Hanks buffer, cells were collected and lysed. Protein was extracted using radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology, Haimen, China) and measured using a bicinchoninic acid (BCA) protein assay kit (Pierce; Thermo Fisher Scientific, Inc.). Protein (~50 µg/lane) was separated on 10% SDS-PAGE gels and transferred to polyvinylidene fluoride membranes. Membranes were then blocked with 5% fat-free milk in TBST buffer [20 mmol/l Tris-HCl (pH 7.5); 150 mmol/l NaCl and 0.05% Tween 20] and subsequently incubated with the following primary antibodies at 4°C overnight: Anti-cleaved caspase-3 (cat. no. 9661S; Cell Signaling Technology, Inc., Danvers, MA, USA), anti-Bcl2-associated X (Bax; cat. no. BS90120; Bioworld Technology, Inc., St Louis Park, MN, USA), anti-B cell lymphoma-2 (Bcl-2; cat. no. BS1511; Bioworld Technology, Inc., St Louis Park, MN, USA), anti-full length caspase-3 (cat. no. sc-56053) and polyclonal anti-β-actin (cat. no. sc-47778; both Santa Cruz Biotechnology, Inc., Dallas, TX, USA) antibodies. Each primary antibody was diluted in Tris-buffered saline with Tween 20 to 1:1,000. The mixture was washed and then incubated at room temperature for 1 h with horseradish peroxidase-conjugated immunoglobulin G secondary antibody (dilution 1:500; cat. no. 074-1506; KPL Inc., Gaithersburg, MD, USA). Membranes were developed using an ECL kit (Pierce; Thermo Fisher Scientific, Inc.) and band quantification was performed via densitometry using a gel image analysis system (GelDoc-It; UVP, LLC, Upland, CA, USA) and GeneSnap software (version 7.8; SynGene, Cambridge, UK). β-actin served as the loading control. Each experiment was repeated three times and three independent experiments were performed.

Statistical analysis

Data were presented as the mean ± standard deviation. One-way analysis of variance was used to assess multiple differences, followed by a Tukey's post-hoc test with SPSS 19.0 software (IBM Corp., Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of HG and liraglutide on cardiomyocyte viability

Cardiomyocyte viability was assessed using an MTT assay, the results of which are presented in Fig. 1. Compared with the NG group, cell viability was significantly decreased in the HG group (P<0.01). Liraglutide treatment (10, 100 and 1,000 nmol/l) significantly improved cell viability following exposure to HG (P<0.05 and P<0.01). However, no significant differences in cell viability were determined between the concentrations of 100 and 1,000 nmol/l liraglutide. Therefore, the current study selected 10 and 100 nmol/l liraglutide for the following experiments. All concentrations of liraglutide (10, 100 and 1,000 nmol/l) did not affect cell viability when exposed to NG, which indicates that liraglutide treatment is not cytotoxic to cardiomyocytes.

Effect of HG and liraglutide on cardiomyocyte apoptosis

The early apoptosis rate of cardiomyocytes was measured (Fig. 2). Compared with the NG group, increased early apoptosis was observed in the presence of HG (P<0.01). However, liraglutide treatment (10 and 100 nmol/l) significantly suppressed the HG-induced increase of early apoptosis (P<0.01). No significant difference in early apoptosis between the OSM and NG groups was identified, indicating that the effect of HG on myocyte apoptosis is independent of high osmotic pressure.

Effect of HG and liraglutide on oxidative stress

MDA content, a classic marker of oxidative damage and SOD activity, a marker of anti-oxidants, were measured. Compared with the NG group, MDA content in the HG group was significantly increased (P<0.01). In contrast, SOD activity was decreased in the HG group compared with the NG group (P<0.01). Treatment with liraglutide markedly decreased the HG-induced increase in MDA content and enhanced SOD activity (P<0.05 and P<0.01). No significant differences in MDA level and SOD activity were identified between the OSM and NG groups (Fig. 3).

Effect of HG and liraglutide on apoptosis-associated proteins

It is well known that apoptosis-associated proteins regulate the progression of apoptosis. Thus, the protein expression of Bax, Bcl-2, cleaved caspase-3 and full length caspase-3 were determined (Figs. 4 and 5, respectively). Cleaved caspase-3/full length caspase-3 was deemed to represent active caspase-3 levels. Compared with the NG group, Bax and active caspase-3 expression were significantly increased and Bcl-2 was markedly decreased in the HG group (P<0.01). Following treatment with liraglutide, Bax and active caspase-3 protein levels were significantly decreased and Bcl-2 was significantly increased when compared with the HG group (P<0.01). No significant differences in the OSM and NG groups were identified.

Discussion

In addition to its glucose-lowering effect, GLP-1 analogs exhibit potential clinical and cardioprotective effects. Arturi et al (20) revealed that treatment with liraglutide improved left ventricular function in patients with type 2 diabetes and a history of post-ischemic chronic heart failure. The Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results-A Long Term Evaluation clinical trial also demonstrated that, among patients with type 2 diabetes who were at high risk for cardiovascular events and were receiving standard therapy, those in the liraglutide group exhibited lower rates of cardiovascular events and mortality from any cause compared with those in the placebo group (11). Furthermore, Okada et al (21) demonstrated that treatment with liraglutide induced a reduction in reactive oxygen markers in patients with type 2 diabetes, hypothesizing that the cardioprotective action of liraglutide may be associated with the alleviation of oxidative stress. It has also been revealed that liraglutide increases the activity of nitric oxide synthase in human endothelial cells, improving their vascular endothelial function (22,23). These cardioprotective actions may be associated with the pleiotropic effects that liraglutide exerts on the heart.

Accumulating evidence has revealed that long-term exposure to HG results in oxidative stress and cardiomyocyte apoptosis, which serve important roles in the pathogenesis of DCM (2426). Consistent with these observations, the results of the present study demonstrated that HG augmented oxidative stress and concurrently triggered the apoptosis signaling pathway, leading to the upregulation of the pro-apoptotic protein Bax and the downregulation of the anti-apoptotic protein Bcl-2. It has been previously reported that the GLP-1 receptor (GLP-1R) agonist, exenatide, attenuates extracellular matrix remodeling, cardiomyocyte hypertrophy and apoptosis in experimental models of type 1 and type 2 diabetes via various mechanisms, including the suppression of oxidative stress and myocardial inflammation, as well as the regulation of endoplasmic reticulum (ER) stress and microvascular barrier function (2729). Noyan-Ashraf et al (9) revealed that treatment with liraglutide reduced infarct development and improved cardiac output in murine models of type 2 diabetes with myocardial infarction (MI) compared with mice treated with metformin, and that the effects of liraglutide on enhanced survival following MI in diabetic mice were independent of glycemic control and weight loss. Their further experiment revealed that liraglutide activated cytoprotective pathways, upregulated the expression of cardioprotective genes (including protein kinase B, glycogen synthase kinase 3β and nuclear factor erythroid factor 2-related factor 2) and inhibited the activation of caspase-3 in diabetic murine hearts, which was an effect that was superior to that of metformin (18). Additionally, Liu et al (16) revealed that liraglutide protects against DCM by inhibiting the ER stress pathway in rat models of type 2 diabetes and that the improvement of cardiac function by liraglutide was independent of glucose control. Inoue et al (17) also demonstrated that liraglutide prevents cardiac oxidative stress and apoptosis by activating the AMP-activated protein kinase (AMPK)-sirtuin 1 (Sirt1) pathway in streptozotocin-induced diabetic rats in vivo. These previous studies confirm that liraglutide inhibits cardiac oxidative stress and protects against DCM in diabetic animals in vivo. To further elucidate the protective mechanism of liraglutide against cardiomyocytes, it is necessary to perform an in vitro study. The present study demonstrated that liraglutide alleviates HG-induced oxidative stress and cardiomyocyte apoptosis, which may be attributable, in part, to the inhibition of Bax expression, the inhibition of caspase-3 activation and the upregulation of Bcl-2 expression. These results are congruent with those of diabetic in vivo models utilized in previous studies. Inoue et al (17) hypothesized that the beneficial effect of liraglutide on diabetic hearts may be associated with the improvement of myocardial fatty acid metabolism in vivo by activating the AMPK-Sirt1 pathway. The results of the current study revealed that liraglutide exhibited a direct preventive effect on cardiomyocyte apoptosis in vitro. However, elucidating the mechanisms by which liraglutide exerts cardioprotection is challenging, as GLP-1R is largely expressed in atrial and not ventricular cardiomyocytes (30,31). Noyan-Ashraf et al (9) determined that liraglutide increased cyclic AMP formation and reduced cardiomyocyte caspase-3 activation in a GLP-1R-dependent manner. The previous study revealed that liraglutide provides cardioprotection and increased survival in GLP-1R CM−/− mice, that liraglutide improved cardiac function in a GLP-1R-independent manner and that atrial GLP-1R is not required for GLP-1R agonist-mediated cardioprotection (32). Therefore, the cardioprotective effects of liraglutide may be mediated through GLP-1R-dependent and GLP-1R-independent pathways (33). Younce et al (34) determined that exendin-4 attenuates HG-induced cardiomyocyte apoptosis in neonatal rat ventricular myocytes in vitro, and that the protective effect is dependent on the inhibition of ER stress, which is downstream of oxidative stress but independent of reduced oxidative stress. However, these differences among previous studies on the cardioprotective actions of GLP-1 analogs may be associated with the different types of GLP-1 analogs used (17,34).

Clinical trials have confirmed that liraglutide exerts anti-oxidative, anti-atherosclerotic and beneficial cardiovascular effects in patients with diabetes (57,1315). Consistent with these data, the present study indicated that liraglutide exerts a cardioprotective effect. The results may reveal one of the mechanisms that underlie the cardiovascular benefit of diabetic patients treated with liraglutide. The present study has certain limitations. There is an absence of data on the effect of liraglutide on myocardial apoptosis in an in vivo rat model of type 2 diabetes. However, a previous study has demonstrated that liraglutide inhibits cardiac myocyte apoptosis by decreasing ER stress in DCM rats (16). Furthermore, although early apoptosis rates and cell viabilities were determined via reliable methods (flow cytometry and cell viability, respectively) (35,36), terminal deoxynucleotidyl-tranferase-mediated dUTP nick end labeling or DNA laddering would have provided stronger evidence to support conclusions. Additionally, the association between oxidative stress and cardiomyocyte apoptosis was not assessed in the present study. Thus, further experimental confirmation is required.

In conclusion, the current study revealed that HG augments oxidative stress and apoptosis in neonatal rat cardiomyocytes. It also demonstrated that liraglutide suppresses HG-induced oxidative stress and cardiomyocyte apoptosis, indicating that the anti-apoptotic actions of liraglutide may be, in part, due to the inhibition of Bax, the inhibition of caspase-3 activation and the upregualtion of Bcl-2.

Acknowledgements

The authors would like to thank Dr Wenjian Li (Department of immunology, School of Basic Medicine, Hebei Medical University) for providing technical assistance.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Author contributions

ZL and LC cultured cardiomyocytes and wrote the manuscript. ZQ and LN performed the superoxide dismutase and malondialdehyde measurements, and western blotting. ZH designed the current study and performed statistical analysis. All authors read and approved the final version of the manuscript.

Ethical approval and consent to participate

The animal protocol was reviewed and approved by the Laboratory Animal Ethical and Welfare Committee of Hebei Medical University (Shijiazhuang, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Jia G, Whaley-Connell A and Sowers JR: Diabetic cardiomyopathy: A hyperglycaemia-and insulin-resistance-induced heart disease. Diabetologia. 61:21–28. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Aneja A, Tang WH, Bansilal S, Garcia MJ and Farkouh ME: Diabetic cardiomyopathy: Insights into pathogenesis, diagnostic challenges, and therapeutic options. Am J Med. 121:748–757. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Wang Y, Sun W, Du B, Miao X, Bai Y, Xin Y, Tan Y, Cui W, Liu B, Cui T, et al: Therapeutic effect of MG-132 on diabetic cardiomyopathy is associated with its suppression of proteasomal activities: Roles of Nrf2 and NF-κB. Am J Physiol Heart Circ Physiol. 304:H567–H578. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Guan SJ, Ma ZH, Wu YL, Zhang JP, Liang F, Weiss JW, Guo QY, Wang JY, Ji ES and Chu L: Long-term administration of fasudil improves cardiomyopathy in streptozotocin-induced diabetic rats. Food Chem Toxicol. 50:1874–1882. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Scheen AJ: Cardiovascular effects of new oral glucose-lowering agents: DPP-4 and SGLT-2 inhibitors. Circ Res. 122:1439–1459. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Drucker DJ: The biology of incretin hormones. Cell Metab. 3:153–165. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Drucker DJ, Habener JF and Holst JJ: Discovery, characterization, and clinical development of the glucagon-like peptides. J Clin Invest. 127:4217–4227. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Drucker DJ: The cardiovascular biology of glucagon-like peptide-1. Cell Metab. 24:15–30. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Noyan-Ashraf MH, Momen MA, Ban K, Sadi AM, Zhou YQ, Riazi AM, Baggio LL, Henkelman RM, Husain M and Drucker DJ: GLP-1R agonist liraglutide activates cytoprotective pathways and improves outcomes after experimental myocardial infarction in mice. Diabetes. 58:975–983. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Dokken BB, La Bonte LR, Davis-Gorman G, Teachey MK, Seaver N and McDonagh PF: Glucagon-like peptide-1 (GLP-1), immediately prior to reperfusion, decreases neutrophil activation and reduces myocardial infarct size in rodents. Horm Metab Res. 43:300–305. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, et al: Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 375:311–322. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, et al: Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 375:1834–1844. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Nauck MA, Meier JJ, Cavender MA, Abd El Aziz M and Drucker DJ: Cardiovascular actions and clinical outcomes with glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Circulation. 136:849–870. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Rizzo M, Abate N, Chandalia M, Rizvi AA, Giglio RV, Nikolic D, Marino Gammazza A, Barbagallo I, Isenovic ER, Banach M, et al: Liraglutide reduces oxidative stress and restores heme oxygenase-1 and ghrelin levels in patients with type 2 diabetes: A prospective pilot study. J Clin Endocrinol Metab. 100:603–606. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Rizzo M, Rizvi AA, Patti AM, Nikolic D, Giglio RV, Castellino G, Li Volti G, Caprio M, Montalto G, Provenzano V, et al: Liraglutide improves metabolic parameters and carotid intima-media thickness in diabetic patients with the metabolic syndrome: An 18-month prospective study. Cardiovasc Diabetol. 15:1622016. View Article : Google Scholar : PubMed/NCBI

16 

Liu J, Liu Y, Chen L, Wang Y and Li J: Glucagon-like peptide-1 analog liraglutide protects against diabetic cardiomyopathy by the inhibition of the endoplasmic reticulum stress pathway. J Diabetes Res. 2013:6305372013. View Article : Google Scholar : PubMed/NCBI

17 

Inoue T, Inoguchi T, Sonoda N, Hendarto H, Makimura H, Sasaki S, Yokomizo H, Fujimura Y, Miura D and Takayanagi R: GLP-1 analog liraglutide protects against cardiac steatosis, oxidative stress and apoptosis in streptozotocin-induced diabetic rats. Atherosclerosis. 240:250–259. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Noyan-Ashraf MH, Shikatani EA, Schuiki I, Mukovozov I, Wu J, Li RK, Volchuk A, Robinson LA, Billia F, Drucker DJ and Husain M: A glucagon-like peptide-1 analog reverses the molecular pathology and cardiac dysfunction of a mouse model of obesity. Circulation. 127:74–85. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Liu C, Xue R, Wu D, Wu L, Chen C, Tan W, Chen Y and Dong Y: REDD1 attenuates cardiac hypertrophy via enhancing autophagy. Biochem Biophys Res Commun. 454:215–220. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Arturi F, Succurro E, Miceli S, Cloro C, Ruffo M, Maio R, Perticone M, Sesti G and Perticone F: Liraglutide improves cardiac function in patients with type 2 diabetes and chronic heart failure. Endocrin. 57:464–473. 2017. View Article : Google Scholar

21 

Okada K, Kotani K, Yaqyu H, Ando A, Osuqa J and Ishibashi S: Effects of treatment with liraglutide on oxidative strss and cardiac natriuretic peptide levels in patients with type 2 diabetes mellitus. Endocrine. 47:962–964. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Dai Y, Mehta JL and Chen M: Glucagon-like peptide-1 receptor agonist liraglutide inhibits endothelin-1 in endothelial cell by repressing nuclear factor-kappa B activation. Cardiovasc Drugs Ther. 27:371–380. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Nandy D, Johnson C, Basu R, Joyner M, Brett J, Svendsen CB and Basu A: The effect of liraglutide on endothelial function in patients with type 2 diabetes. Diab Vasc Dis Res. 11:419–430. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Guo S, Yao Q, Ke Z, Chen H, Wu J and Liu C: Resveratrol attenuates high glucose-induced oxidative stress and cardiomyocyte apoptosis through AMPK. Mol Cell Endocrinol. 412:85–94. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Zhang F, Lin X, Yu L, Li W, Qian D, Cheng P, He L, Yang H and Zhang C: Low-dose radiation prevents type 1 diabetes-induced cardiomyopathy via activation of AKT mediated anti-apoptotic and anti-oxidant effects. J Cell Mol Med. 20:1352–1366. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Despa S, Margulies KB, Chen L, Knowlton AA, Havel PJ, Taegtmeyer H, Bers DM and Despa F: Hyperamylinemia contributes to cardiac dysfunction in obesity and diabetes: A study in humans and rats. Circ Res. 110:598–608. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Monji A, Mitsui T, Bando YK, Aoyama M, Shigeta T and Murohara T: Glucagon-like peptide-1 receptor activation reverses cardiac remodeling via normalizing cardiac steatosis and oxidative stress in type 2 diabetes. Am J Physiol Heart Circ Physiol. 305:H295–H304. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Wang D, Luo P, Wang Y, Li W, Wang C, Sun D, Zhang R, Su T, Ma X, Zeng C, et al: Glucagon-like peptide-1 protects against cardiac microvascular injury in diabetes via a cAMP/PKA/Rho-dependent mechanism. Diabetes. 62:1697–1708. 2013. View Article : Google Scholar : PubMed/NCBI

29 

XiaoTian L, QiNan W, XiaGuang G, WuQuan D, Bing C and ZiWen L: Exenatide activates the APPL1-AMPK-PPARα axis to prevent diabetic cardiomyocyte apoptosis. J Diabetes Res. 2016:42197352016. View Article : Google Scholar : PubMed/NCBI

30 

Kim M, Platt MJ, Shibasaki T, Quaggin SE, Backx PH, Seino S, Simpson JA and Drucker DJ: GLP-1 receptor activation and Epac2 link atrial natriuretic peptide secretion to control of blood pressure. Nat Med. 19:567–575. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Richards P, Parker HE, Adriaenssens AE, Hodgson JM, Cork SC, Trapp S, Gribble FM and Reimann F: Identification and characterisation of glucagon-like peptide-1 receptor expressing cells using a new transgenic mouse model. Diabetes. 63:1224–1233. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Ussher JR, Baggio LL, Campbell JE, Mulvihill EE, Kim M, Kabir MG, Cao X, Baranek BM, Stoffers DA, Seeley RJ and Drucker DJ: Inactivation of the cardiomyocyte glucagon-like peptide-1 receptor (GLP-1R) unmasks cardiomyocyte-independent GLP-1R-mediated cardioprotection. Mol Metab. 3:507–517. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Ban K, Noyan-Ashraf MH, Hoefer J, Bolz SS, Drucker DJ and 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. View Article : Google Scholar : PubMed/NCBI

34 

Younce CW, Burmeister MA and Ayala JE: Exendin-4 attenuates high glucose-induced cardiomyocyte apoptosis via inhibition of endoplasmic reticulum stress and activation of SERCA2a. Am J Physiol Cell Physiol. 304:C508–C518. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Liu H, Chen X, Han Y, Li C, Chen P, Su S, Zhang Y and Pan Z: Rho kinase inhibition by fasudil suppresses lipopolysaccharide-induced apoptosis of rat pulmonary microvascular endothelial cells via JNK and p38 MAPK pathway. Biomed Pharmacother. 68:267–275. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Gao H, Hou F, Dong R, Wang Z, Zhao C, Tang W and Wu Y: Rho-Kinase inhibitor fasudil suppresses high glucose-induced H9c2 cell apoptosis through activation of autophagy. Cardiovasc Ther. 34:352–359. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2019
Volume 17 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang L, Li C, Zhu Q, Li N and Zhou H: Liraglutide, a glucagon‑like peptide‑1 analog, inhibits high glucose‑induced oxidative stress and apoptosis in neonatal rat cardiomyocytes. Exp Ther Med 17: 3734-3740, 2019
APA
Zhang, L., Li, C., Zhu, Q., Li, N., & Zhou, H. (2019). Liraglutide, a glucagon‑like peptide‑1 analog, inhibits high glucose‑induced oxidative stress and apoptosis in neonatal rat cardiomyocytes. Experimental and Therapeutic Medicine, 17, 3734-3740. https://doi.org/10.3892/etm.2019.7388
MLA
Zhang, L., Li, C., Zhu, Q., Li, N., Zhou, H."Liraglutide, a glucagon‑like peptide‑1 analog, inhibits high glucose‑induced oxidative stress and apoptosis in neonatal rat cardiomyocytes". Experimental and Therapeutic Medicine 17.5 (2019): 3734-3740.
Chicago
Zhang, L., Li, C., Zhu, Q., Li, N., Zhou, H."Liraglutide, a glucagon‑like peptide‑1 analog, inhibits high glucose‑induced oxidative stress and apoptosis in neonatal rat cardiomyocytes". Experimental and Therapeutic Medicine 17, no. 5 (2019): 3734-3740. https://doi.org/10.3892/etm.2019.7388