Dipeptidyl peptidase‑4 inhibitor sitagliptin prevents high glucose‑induced apoptosis via activation of AMP‑activated protein kinase in endothelial cells

  • Authors:
    • Chao Wu
    • Shunying Hu
    • Nanping Wang
    • Jianwei Tian
  • View Affiliations

  • Published online on: April 21, 2017     https://doi.org/10.3892/mmr.2017.6501
  • Pages: 4346-4351
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Diabetes mellitus (DM), which is a chronic metabolic disorder, is the primary risk factor of life‑threatening vascular complications. Endothelial apoptosis is important in the development of the initial vascular lesion preceding the diabetic disease. Sitagliptin is a dipeptidyl peptidase‑4 (DPP‑4) inhibitor and extensively used in the clinical treatment of DM. DPP‑4 inhibitors have been demonstrated to be beneficial in the improvement of endothelial homeostasis, however the molecular mechanism by which they exhibit these effects remains to be elucidated. The effect of sitagliptin on endothelial apoptosis was examined in cultured human umbilical vein endothelial cells (HUVECs) incubated with high glucose (HG). The present study demonstrated that treatment of HUVECs with HG increased reactive oxygen species (ROS) production, stimulated mitochondrial depolarization and resulted in cell apoptosis. Pretreatment of HUVECs with sitagliptin significantly prevented HG‑induced endothelial apoptosis. It was further demonstrated that sitagliptin effectively inhibited ROS generation and mitochondrial membrane potential collapse. Similarly, adenosine monophosphate‑activated protein kinase (AMPK) activation by sitagliptin protected against HG‑induced ROS production, mitochondrial membrane potential collapse and endothelial cell apoptosis, as detected via western blotting and flow cytometry analysis. The present study therefore revealed a novel mechanism of sitagliptin‑mediated AMPK activation in preventing endothelial apoptosis and indicated the therapeutic potential of sitagliptin in vascular complications associated with endothelial apoptosis.

Introduction

Diabetes mellitus (DM) is an increasingly prevalent worldwide disease that is challenging human health and is currently of primary concern. As one of the major risk factors for cardiovascular diseases, type 2 diabetes contributes greatly to the occurrence of disabilities in later life and to mortality (1). Over 50% of mortality events resulting from type 2 diabetes are attributable to cardiovascular diseases (2), including stroke and myocardial infarction. Microvascular endothelial apoptosis is important in the development of the initial vessel lesions of vascular complications in DM (3).

Dipeptidyl peptidase-4 (DPP-4) inhibitors are of a class of oral hypoglycemic agents, which reduce blood glucose levels with a low risk of hypoglycemia and weight gain. DPP-4 is an enzyme expressed on blood vessels, myocardium and myeloid cells and is responsible for the inactivation of endogenous glucoregulatory peptides, termed incretins (4). Glucagon-like peptide 1 (GLP-1) and gastric inhibitory peptide are two well-studied incretins. GLP-1 prolongs gastric emptying, reduces appetite, inhibits glucagon release and stimulates insulin in a glucose-dependent manner. GLP-1 receptor (GLP-1R) agonists have been used in the treatment of patients with type 2 diabetes (5). Sitagliptin was the first clinically used DPP-4 inhibitor and was approved by the US Food and Drug Administration for the treatment of type 2 diabetes in 2006 (6). Recent studies in apolipoprotein E-deficient mice revealed that sitagliptin improved endothelial dysfunction, enhanced endothelial nitric oxide synthase (eNOS) phosphorylation (7) and reduced the atherosclerotic plaque area (8), suggesting that DPP-4 inhibitors may have further potential therapeutic effects beyond the incretin-dependent hypoglycemic action. DPP-4 inhibitors have been demonstrated to exhibit cardiovascular protective functions, however their effect on endothelial apoptosis and the underlying mechanism in diabetes remains to be fully elucidated.

Adenosine monophosphate-activated protein kinase (AMPK) is a cellular energy and stress sensor (9). In diabetes, AMPK has been observed to be dephosphorylated and inactive (10). A previous study revealed that AMPK activation significantly prevents the oxidative stress-induced apoptosis of human umbilical vein endothelial cells (HUVECs) (11). Various studies have additionally demonstrated that AMPK prevents apoptosis via inhibiting reactive oxygen species (ROS) generation by mitochondria (12) and nicotinamide adenine dinucleotide phosphate [NAD(P)H] oxidase (13). The molecular mechanism regarding how the DPP-4 inhibitor regulates endothelial homeostasis and the associated functional role of AMPK remains to be elucidated. The present study aimed to verify the mechanism by which the DPP-4 inhibitor sitagliptin protects against high glucose (HG)-induced vascular endothelial cell apoptosis and examine if AMPK is involved in this regulatory process.

Materials and methods

Reagents

Sitagliptin (phosphate) was provided by Cayman Chemical Company (Ann Arbor, MI, USA) and 5-aminoimidazole-4-carboxamide riboside (AICAR) was purchased from Beyotime Institute of Biotechnology (Haimen, China). Compound C was obtained from Sigma-Aldrich; Merck KGaA (Darmstadt, Germany). Monoclonal rabbit anti-phospho-AMPKα antibody (catalog no. 2535p) and anti-AMPK antibody (catalog no. 2603p) were obtained from Cell Signaling Technology, Inc. (Danvers, MA, USA). The following antibodies were also used: Monoclonal mouse anti-β-actin antibody (catalog no. sc-47778) and horseradish peroxidase-conjugated goat anti-rabbit/mouse secondary antibody (catalog no. sc-2004/sc-2005) from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA).

Cell culture

HUVECs were isolated by collagenase digestion from fresh cord umbilical veins, as previously described (14). The flesh cord umbilical veins were obtained from normal cesarean section surgery. This was approved by Air Force General Hospital ethics committee with informed written consent. HUVECs between passages 3 and 6 were cultured in endothelial cell medium (ScienCell Research Laboratories, Inc., Carlsbad, CA, USA) containing basal medium, supplemented with 5% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and 1% endothelial cell growth supplement with antibiotics (100 U/ml penicillin G and 100 µg/ml streptomycin sulfate). This was conducted in a humidified atmosphere containing 5% CO2, at 37°C.

Western blotting

To determine the effect of sitagliptin on AMPK activation, the HUVECs were treated with 1 µM sitagliptin for 0.5, 1, 2 and 4 h or 100 µM AMPK activator AICAR, for 0.5 h. To detect the inhibitory effect of the AMPK inhibitor compound C on sitagliptin-induced AMPKα phosphorylation, HUVECs were incubated with 1 µM sitagliptin, 10 µM compound C or 1 µM sitagliptin plus 10 µM compound C for 2 h. The cytoplasmic protein of cells was extracted with ice-cold hypotonic lysis buffer [50 mM Tris-HCl, pH 7.5, 15 mM EGTA, 0.1% (vol/vol) Triton X-100, 100 mM NaCl and complete protease inhibitor cocktail] as previously described (15). Cell lysates were first snap frozen in liquid nitrogen and then centrifuged at 12,000 × g at 4°C for 20 min, for collection of the supernatant. Protein concentration was measured using the BCA method. Equal amounts of protein (10 µg per sample) were separated on 10% sodium dodecyl sulfate-polyacrylamide gels electrophoresis and blotted onto polyvinylidene difluoride membranes. Following incubation with no fat milk at 25°C for 20 min, the membranes were reacted with anti-phospho-AMPKα antibody (1:1,000) and anti-AMPKα antibody (1:1,000) at 4°C overnight, then reacted with appropriate horseradish peroxidase-conjugated secondary antibodies (1:3,000) for 2 h at 25°C. Proteins were visualized with an enhanced chemiluminescence kit, as previously described (16). Densitometry analysis was performed for three independent experiments using the Image J Gel Analysis tool (National Institutes of Health, Bethesda, MD, USA).

Measurement of endothelial apoptosis

HUVECs (1×105) were incubated with HG (33 mM) in the presence of 1 µM sitagliptin, 100 µM AICAR or 1 µM sitagliptin plus 10 µM AMPK inhibitor compound C for 48 h. Induction of apoptosis in the treated groups was assessed by Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) double staining detection kit (Nanjing KeyGen Biotech Co., Ltd., Nanjing, China), according to the manufacturer's protocol. Briefly, cells were incubated with 33 mM D-glucose in the presence of the aforementioned agents for 48 h and gently digested with 1 ml 0.25% trypsin (Thermo Fisher Scientific, Inc.) for 2 min. The trypsinized cells were washed once with endothelial cell medium containing 5% fetal bovine serum prior to collection by centrifugation at 1,000 × g and room temperature for 20 min. Cells were resuspended in 500 µl of 1X binding buffer, followed by incubation with 5 µl of Annexin V-FITC and 5 µl of PI (50 µg/ml) for 10 min in the dark. Binding buffer, Annexin V-FITC and PI are components of the detection kit. All procedures subsequent to cell incubation were performed at room temperature. Stained cells were monitored by flow cytometry (BD FACSCalibur; BD Biosciences, San Jose, CA, USA) and analyzed with BD FACSDiva software (version 6.0; BD Biosciences).

Measurement of ROS generation

A ROS-specific fluorescent probe, 2′,7′-dichlorodihydrofluorescein diacetate (H2DCFDA; Molecular Probes; Thermo Fisher Scientific, Inc.) was used for the measurement of cytosolic ROS production. HUVECs were incubated with 33 mM D-glucose in the presence of 1 µM sitagliptin, 100 µM AICAR or 1 µM sitagliptin plus 10 µM compound C for 48 h, then cells were stained with 10 µM H2DCFDA fluorescent probe in serum-free endothelial cell medium at 37°C for 30 min. The labeled cells were then washed twice with serum-free endothelial cell medium and the levels of ROS were immediately analyzed by flow cytometry (BD FACSCalibur; BD Biosciences).

Mitochondrial membrane potential (ΔΨm) assay

To measure ΔΨm, HUVECs were treated with 1 µM sitagliptin, 100 µM AICAR or 1 µM sitagliptin plus 10 µM compound C prior to exposure to 33 mM D-glucose for 48 h. Following incubation, cells were collected and stained with 2 µM ΔΨm-specific fluorescent dye JC-1 (Molecular Probes; Thermo Fisher Scientific, Inc.) at 37°C in an atmosphere containing 5% CO2, for 20 min. Flow cytometry (BD FACSCalibur; BD Biosciences) was used to detect ΔΨm for each treatment group. JC-1 accumulates in mitochondria in a potential-dependent manner. In normal mitochondria with high ΔΨm, JC-1 aggregates with red fluorescence. In apoptotic cells with injured mitochondria membrane, JC-1 alters to monomers, and emits green fluorescence. ΔΨm is determined by red/green fluorescence intensity ratio.

Statistical analysis

Data are expressed as the mean ± standard error of the mean. One-way analysis of variance was used to determine differences among the mean values of treatments. SPSS software, version 20.0 (IBM SPSS, Armonk, NY, USA) was used for the statistical data analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

Sitagliptin prevents HG-induced endothelial apoptosis

The present study examined the effect of sitagliptin on HUVECs incubated with HG. Cell apoptosis of the pretreated groups was measured by Annexin V-FITC/PI double staining and monitored by flow cytometry (Fig. 1A). It was observed that HG significantly increased cell apoptosis, and this HG-induced endothelial cell apoptosis was prevented by sitagliptin or the AMPK activator, AICAR. However, compound C, an AMPK inhibitor, reversed the inhibition of apoptosis by sitagliptin (Fig. 1B). This therefore indicated that AMPK is important in the regulatory action of sitagliptin.

Sitagliptin activates AMPKα phosphorylation in vascular endothelial cells

As AMPK was observed to be involved in sitagliptin-mediated prevention of endothelial cell apoptosis, the present study aimed to determine the effect of sitagliptin on AMPK activity. HUVECs were incubated with 1 µM sitagliptin at different times ranging from 0.5–4 h. Phosphorylation of AMPKα (p-AMPKα) was determined by western blotting (Fig. 2A). Sitagliptin stimulated AMPKα (Thr172) phosphorylation from 2 h, and this phosphorylation activity prevailed until 4 h. AICAR enhanced AMPK phosphorylation in endothelial cells in a similar manner to sitagliptin, following incubation with the cells for 0.5 h (Fig. 2B). The effect of compound C on sitagliptin-induced AMPKα phosphorylation was additionally examined (Fig. 2C). As presented in (Fig. 2D), sitagliptin-stimulated AMPKα activation was significantly inhibited by compound C. These findings suggested that sitagliptin induces AMPKα phosphorylation.

Sitagliptin decreases high glucose-induced ROS generation

In vascular endothelial cells, the hyperglycemia load increases generation of ROS (17), which subsequently contributes to cell apoptosis. To observe the effect of sitagliptin pretreatment on HG-induced cytosolic ROS generation, cytosolic ROS levels were detected via flow cytometry (Fig. 3A). It was observed that high glucose significantly increased ROS production, however this was suppressed with pretreatment with 1 µM sitagliptin. In addition, AICAR effectively inhibited generation of ROS, whereas compound C diminished the inhibitory effect of sitagliptin (Fig. 3B). These data suggested that sitagliptin inhibits cytosolic ROS via AMPK activation. The effect of sitagliptin on ROS-mediated mitochondrial dysfunction, under conditions of hyperglycemia were then examined.

Sitagliptin restores the loss of ΔΨm

ROS-mediated ΔΨm collapse was previously demonstrated to initiate mitochondrial-dependent apoptosis in DM (18). The present study proceeded to characterize HG-induced ΔΨm alterations and examine if sitagliptin protects against ΔΨm collapse. JC-1 staining detection by flow cytometry was performed. Mitochondrial depolarization is determined by a decrease in aggregate/monomer fluorescence ratio (Fig. 4A). In a similar manner to that exhibited by AICAR, 1 µM sitagliptin restored HG-induced ΔΨm collapse, and this effect was blocked by compound C (Fig. 4B). These results suggested that AMPK is important in the regulatory actions of sitagliptin in HG-induced endothelial apoptosis.

Discussion

The present study demonstrated that the DPP-4 inhibitor, sitagliptin, functions as a regulator of endothelial cell apoptosis. In HUVECs, sitagliptin effectively prevented HG-induced apoptosis. The underlying mechanisms may involve inhibition of ROS and the downstream ΔΨm collapsing pathway, induced via AMPKα activation, as summarized in Fig. 5. Overall, the present study has reported novel findings, suggesting DPP-4 inhibitor-mediated AMPK activation as a therapeutic target for vascular endothelial apoptosis.

Diabetes is a major risk factor for cardiovascular disease. The risk of the development of cardiovascular complications in diabetes suggests a need for further therapeutic treatments, which may modulate disease-specific mechanisms, including endothelial apoptosis. DPP-4 inhibitors, including sitagliptin, alogliptin and vildagliptin are safe, well-tolerated hypoglycemic agents that have exhibited beneficial therapeutic effects in diabetes. Previous studies have demonstrated that DPP-4 inhibitors have substantial implications in the cardiovascular system. Alogliptin relaxes reconstructed aortic segments (19), and incubation of HUVECs with vildagliptin has been demonstrated to result in phosphorylation of eNOS and serine/threonine kinase 1, increasing nitric oxide synthesis (20). Various animal model studies have demonstrated beneficial effects of DPP-4 inhibitors in improving blood pressure and endothelial dysfunction (7,21). The present study revealed the novel mechanism of DPP-4 inhibitor-mediated apoptosis prevention. The results demonstrated that sitagliptin protected against HG-induced apoptosis in HUVECs, an effect additionally exhibited by the AMPK activator AICAR. Compound C, an AMPK inhibitor, diminished the inhibitory effect of the sitagliptin pretreatment. The potential link between sitagliptin and AMPK was then determined. The data demonstrated that AMPKα phosphorylation was activated by sitagliptin and compound C inhibited sitagliptin-induced AMPKα activation. Therefore, it was demonstrated that AMPK activation is important in sitagliptin-mediated protection against HG-induced vascular endothelial apoptosis.

AMPK is composed of catalytic α-subunit and regulatory β- and γ-subunits (9). The activation of AMPK occurs via α-subunit phosphorylation at Thr172 (22). AMPK is an important regulator of metabolic homeostasis, and is considered a therapeutic target for the prevention of diabetic complications (23). Various reports have demonstrated that the AMPK signaling pathway exhibits a protective effect against endothelial dysfunction (24) and prevents apoptosis of HUVECs (11) consistent with the findings of the present study. A previous study reported that AMPKα acts as a physiological suppressor of NAD(P)H oxidase and ROS generation in endothelial cells (12), whereas a further study conversely indicated that AMPK is activated by ROS (25). Hyperglycemia-induced intracellular ROS production and associated downstream mitochondrial fission, stimulated ΔΨm collapse, which resulted in mitochondrial-dependent apoptosis (26). The present study observed a significant increase in cytosolic ROS generation and ΔΨm collapse upon incubation with HG in HUVECs. The ROS production and ΔΨm collapse were suppressed by pretreatment with sitagliptin or AICAR. Compound C reversed the effect of sitagliptin. Therefore, there may be a negative feedback loop between AMPK and ROS, in which ROS generation potentiates AMPK activation, resulting in a further inhibition in intracellular ROS production.

Sitagliptin was demonstrated to prevent endothelial apoptosis via AMPKα activation, therefore it is necessary to elucidate the mechanism by which sitagliptin activates AMPKα. The Ca2+/calmodulin-activated protein kinase kinases (CaMKK), particularly CaMKKβ (27) and the liver kinase B1 (LKB1)-STRAD-MO25 complex (28) are major upstream kinases of AMPK in mammals, and the upstream kinase LKB1 is important for the activation of AMPK by AICAR (29). Sitagliptin mimicked the preventive effect of AICAR on HG-induced ROS production, ΔΨm collapse, and endothelial cell apoptosis, indicating that sitagliptin-mediated AMPK HG-induced AMPKα activation may involve LKB1. It was additionally reported that sitagliptin treatment improved endothelial function in vivo via sequential activation of the LKB1/AMPKα/eNOS axis (30).

In conclusion, the results of the present study indicated that the DPP-4 inhibitor sitagliptin effectively prevented HG-induced cytosolic ROS generation, ΔΨm collapse and apoptosis via activation of AMPKα in endothelial cells. These results suggested sitagliptin may act as a potential novel therapeutic agent to treat vascular complications in diabetes.

Acknowledgements

The present study was supported by The National Natural Science Foundation of China (grant no. 81070209). The authors would like to thank Ms. Guohua Ma (Department of Cardiology, Beijing Tian Tan Hospital, Beijing, China) for providing HUVECs, Dr Zhipeng Wang (Institute of Cardiovascular Science, Peking University, Beijing, China) for helpful discussions and Dr Yahan Liu (Institute of Cardiovascular Science, Peking University) for technical assistance.

Glossary

Abbreviations

Abbreviations:

DPP-4

dipeptidyl peptidase-4

ROS

reactive oxygen species

DM

diabetes mellitus

HG

high glucose

HUVECs

human umbilical vein endothelial cells

ΔΨm

mitochondrial membrane potential

AMPK

adenosine monophosphate-activated protein kinase

References

1 

Mazzone T, Chait A and Plutzky J: Cardiovascular disease risk in type 2 diabetes mellitus: Insights from mechanistic studies. Lancet. 371:1800–1809. 2008. View Article : Google Scholar :

2 

Zhong J, Maiseyeu A, Davis SN and Rajagopalan S: DPP4 in cardiometabolic disease: Recent insights from the laboratory and clinical trials of DPP4 inhibition. Circ Res. 116:1491–1504. 2015. View Article : Google Scholar :

3 

Libby P: Inflammation in atherosclerosis. Nature. 420:868–874. 2002. View Article : Google Scholar

4 

Drucker DJ and 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. View Article : Google Scholar

5 

Ussher JR and Drucker DJ: Cardiovascular biology of the incretin system. Endocr Rev. 33:187–215. 2012. View Article : Google Scholar :

6 

Drucker D, Easley C and Kirkpatrick P: Sitagliptin. Nat Rev Drug Discov. 6:109–110. 2007. View Article : Google Scholar

7 

Matsubara J, Sugiyama S, Sugamura K, Nakamura T, Fujiwara Y, Akiyama E, Kurokawa H, Nozaki T, Ohba K, Konishi M, et al: A dipeptidyl peptidase-4 inhibitor, des-fluoro-sitagliptin, improves endothelial function and reduces atherosclerotic lesion formation in apolipoprotein E-deficient mice. J Am Coll Cardiol. 59:265–276. 2012. View Article : Google Scholar

8 

Zeng Y, Li C, Guan M, Zheng Z, Li J, Xu W, Wang L, He F and Xue Y: The DPP-4 inhibitor sitagliptin attenuates the progress of atherosclerosis in apolipoprotein-E-knockout mice via AMPK- and MAPK-dependent mechanisms. Cardiovasc Diabetol. 13:322014. View Article : Google Scholar :

9 

Hardie DG, Ross FA and Hawley SA: AMPK: A nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol. 13:251–262. 2012. View Article : Google Scholar

10 

Eid AA, Ford BM, Block K, Kasinath BS, Gorin Y, Ghosh-Choudhury G, Barnes JL and Abboud HE: AMP-activated protein kinase (AMPK) negatively regulates Nox4-dependent activation of p53 and epithelial cell apoptosis in diabetes. J Biol Chem. 285:37503–37512. 2010. View Article : Google Scholar :

11 

Ido Y, Carling D and Ruderman N: Hyperglycemia-induced apoptosis in human umbilical vein endothelial cells: Inhibition by the AMP-activated protein kinase activation. Diabetes. 51:159–167. 2002. View Article : Google Scholar

12 

Wang S, Zhang M, Liang B, Xu J, Xie Z, Liu C, Viollet B, Yan D and Zou MH: AMPKalpha2 deletion causes aberrant expression and activation of NAD(P)H oxidase and consequent endothelial dysfunction in vivo: Role of 26S proteasomes. Circ Res. 106:1117–1128. 2010. View Article : Google Scholar :

13 

Colombo SL and Moncada S: AMPKalpha1 regulates the antioxidant status of vascular endothelial cells. Biochem J. 421:163–169. 2009. View Article : Google Scholar

14 

Pan B, Yu B, Ren H, Willard B, Pan L, Zu L, Shen X, Ma Y, Li X, Niu C, et al: High-density lipoprotein nitration and chlorination catalyzed by myeloperoxidase impair its effect of promoting endothelial repair. Free Radic Biol Med. 60:272–281. 2013. View Article : Google Scholar

15 

Mao G, Liu Y, Fang X, Liu Y, Fang L, Lin L, Liu X and Wang N: Tumor-derived microRNA-494 promotes angiogenesis in non-small cell lung cancer. Angiogenesis. 18:373–382. 2015. View Article : Google Scholar

16 

Yin R, Fang L, Li Y, Xue P, Li Y, Guan Y, Chang Y, Chen C and Wang N: Pro-inflammatory Macrophages suppress PPARγ activity in Adipocytes via S-nitrosylation. Free Radic Biol Med. 89:895–905. 2015. View Article : Google Scholar

17 

Paneni F, Mocharla P, Akhmedov A, Costantino S, Osto E, Volpe M, Lüscher TF and Cosentino F: Gene silencing of the mitochondrial adaptor p66(Shc) suppresses vascular hyperglycemic memory in diabetes. Circ Res. 111:278–289. 2012. View Article : Google Scholar

18 

Yee C, Yang W and Hekimi S: The intrinsic apoptosis pathway mediates the pro-longevity response to mitochondrial ROS in C. Elegans. Cell. 157:897–909. 2014. View Article : Google Scholar :

19 

Shah Z, Pineda C, Kampfrath T, Maiseyeu A, Ying Z, Racoma I, Deiuliis J, Xu X, Sun Q, Moffatt-Bruce S, et al: Acute DPP-4 inhibition modulates vascular tone through GLP-1 independent pathways. Vascul Pharmacol. 55:2–9. 2011. View Article : Google Scholar :

20 

Ishii M, Shibata R, Kondo K, Kambara T, Shimizu Y, Tanigawa T, Bando YK, Nishimura M, Ouchi N and Murohara T: Vildagliptin stimulates endothelial cell network formation and ischemia-induced revascularization via an endothelial nitric-oxide synthase-dependent mechanism. J Biol Chem. 289:27235–27245. 2014. View Article : Google Scholar :

21 

Aroor AR, Sowers JR, Bender SB, Nistala R, Garro M, Mugerfeld I, Hayden MR, Johnson MS, Salam M, Whaley-Connell A and Demarco VG: Dipeptidylpeptidase inhibition is associated with improvement in blood pressure and diastolic function in insulin-resistant male Zucker obese rats. Endocrinology. 154:2501–2513. 2013. View Article : Google Scholar :

22 

Oakhill JS, Steel R, Chen ZP, Scott JW, Ling N, Tam S and Kemp BE: AMPK is a direct adenylate charge-regulated protein kinase. Science. 332:1433–1435. 2011. View Article : Google Scholar

23 

Viollet B, Lantier L, Devin-Leclerc J, Hebrard S, Amouyal C, Mounier R, Foretz M and Andreelli F: Targeting the AMPK pathway for the treatment of Type 2 diabetes. Front Biosci (Landmark Ed). 14:3380–3400. 2009. View Article : Google Scholar :

24 

Xu Q and Si LY: Protective effects of AMP-activated protein kinase in the cardiovascular system. J Cell Mol Med. 14:2604–2613. 2010. View Article : Google Scholar :

25 

Hawley SA, Ross FA, Chevtzoff C, Green KA, Evans A, Fogarty S, Towler MC, Brown LJ, Ogunbayo OA, Evans AM and Hardie DG: Use of cells expressing gamma subunit variants to identify diverse mechanisms of AMPK activation. Cell Metab. 11:554–565. 2010. View Article : Google Scholar :

26 

Bhatt MP, Lim YC, Kim YM and Ha KS: C-peptide activates AMPKα and prevents ROS-mediated mitochondrial fission and endothelial apoptosis in diabetes. Diabetes. 62:3851–3862. 2013. View Article : Google Scholar :

27 

Hawley SA, Pan DA, Mustard KJ, Ross L, Bain J, Edelman AM, Frenguelli BG and Hardie DG: Calmodulin-dependent protein kinase kinase-beta is an alternative upstream kinase for AMP-activated protein kinase. Cell Metab. 2:9–19. 2005. View Article : Google Scholar

28 

Shaw RJ, Kosmatka M, Bardeesy N, Hurley RL, Witters LA, DePinho RA and Cantley LC: The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. In: Proc Natl Acad Sci USA. 101. pp. 3329–3335. 2004; View Article : Google Scholar :

29 

Fisslthaler B and Fleming I: Activation and signaling by the AMP-activated protein kinase in endothelial cells. Circ Res. 105:114–127. 2009. View Article : Google Scholar

30 

Liu L, Liu J, Wong WT, Tian XY, Lau CW, Wang YX, Xu G, Pu Y, Zhu Z, Xu A, et al: Dipeptidyl peptidase 4 inhibitor sitagliptin protects endothelial function in hypertension through a glucagon-like peptide 1-dependent mechanism. Hypertension. 60:833–841. 2012. View Article : Google Scholar

Related Articles

Journal Cover

June-2017
Volume 15 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wu C, Hu S, Wang N and Tian J: Dipeptidyl peptidase‑4 inhibitor sitagliptin prevents high glucose‑induced apoptosis via activation of AMP‑activated protein kinase in endothelial cells. Mol Med Rep 15: 4346-4351, 2017
APA
Wu, C., Hu, S., Wang, N., & Tian, J. (2017). Dipeptidyl peptidase‑4 inhibitor sitagliptin prevents high glucose‑induced apoptosis via activation of AMP‑activated protein kinase in endothelial cells. Molecular Medicine Reports, 15, 4346-4351. https://doi.org/10.3892/mmr.2017.6501
MLA
Wu, C., Hu, S., Wang, N., Tian, J."Dipeptidyl peptidase‑4 inhibitor sitagliptin prevents high glucose‑induced apoptosis via activation of AMP‑activated protein kinase in endothelial cells". Molecular Medicine Reports 15.6 (2017): 4346-4351.
Chicago
Wu, C., Hu, S., Wang, N., Tian, J."Dipeptidyl peptidase‑4 inhibitor sitagliptin prevents high glucose‑induced apoptosis via activation of AMP‑activated protein kinase in endothelial cells". Molecular Medicine Reports 15, no. 6 (2017): 4346-4351. https://doi.org/10.3892/mmr.2017.6501