Apelin-13 stimulates angiogenesis by promoting cross‑talk between AMP-activated protein kinase and Akt signaling in myocardial microvascular endothelial cells

  • Authors:
    • Xiaoman Yang
    • Wei Zhu
    • Pu Zhang
    • Kankai Chen
    • Lijie Zhao
    • Jingbo Li
    • Meng Wei
    • Mingya Liu
  • View Affiliations

  • Published online on: February 25, 2014     https://doi.org/10.3892/mmr.2014.1984
  • Pages: 1590-1596
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Currently, there is major interest in the functions of apelin-13, an endogenous ligand for the orphan G-protein coupled receptor APJ, a receptor that closely resembles the angiotensin receptor AT1. In the present study, the role of apelin-13 in angiogenesis and its mechanism as a novel angiogenic factor in myocardial microvascular endothelial cells (MMVECs) was investigated. It was revealed that apelin-13 can promote proliferation, migration and tube formation in MMVECs. In addition, apelin-13 dose dependently stimulated the phosphorylation of AMP-activated protein kinase (AMPK) and endothelial nitric oxide synthase (eNOS) at Thr-172 and Ser-1179, respectively. The treatment with the AMPK (compound C) and protein kinase Akt/protein kinase B (Akt; LY294002) inhibitor significantly suppressed the apelin‑13-induced AMPK, Akt and eNOS phosphorylation. They also inhibited the apelin13‑stimulated endothelial cell migration and tube formation. Therefore, we hypothesize that apelin-13 promotes angiogenesis through the modulation of AMPK and Akt signaling in MMVECs.

Introduction

Apelin is a novel adipocyte-derived factor that binds the orphan G protein-coupled receptor (GPCR) APJ with high affinity (1). Apelin mRNA encodes a 77-amino-acid prepropeptide, which is proteolytically cleaved to yield bioactive peptides of 36, 17 and 13 amino acids (aa) in size. Each of these peptides contains the extreme C-terminal region of the precursor protein and their bioactivity resides in the C-terminal 13 aa fragment (2). The sequence of the 13 aa apelin peptide is highly conserved among different species, suggesting that its critical function has been evolutionarily conserved (3).

The apelinergic system has a widespread pattern of distribution (4,5). However, the majority of studies regarding the function of apelin/APJ are on the cardiovascular system, due to its similarity to the angiotensin receptor. In the cardiovascular system, apelin reduces blood pressure and modulates the contractility of cardiac tissue and blood vessels (6,7,8). Apelin signaling is also associated with tumor angiogenesis and vascular regeneration during embryogenesis (9).

AMP-activated protein kinase (AMPK) is a stress-activated protein kinase, which is involved in the regulation of energy and metabolic homeostasis (10). In vascular endothelium, AMPK signaling is required for vascular endothelial growth factor (VEGF)-induced nitric oxide (NO) production, migration and differentiation under conditions of hypoxia (11). Previous studies indicated that apelin-13 is able to regulate glucose and lipid metabolism (12,13) via the activation of AMPK signaling. The disruption of apelin-13 signaling can increase the hypoxia-induced pulmonary hypertension mediated by decreased activation of AMPK and endothelial nitric oxide synthase (eNOS) (14). In addition, as protein kinase Akt/protein kinase B (Akt) signaling participates in vascular homeostasis and angiogenesis (15,16), it can also phosphorylate eNOS (17), resulting in NO production and subsequently leading to the regulation of vasomotor responses. Thus, it was hypothesized that AMPK and Akt signaling may participate in the regulation of angiogenesis stimulated by apelin-13.

The aim of the present study was to investigate the role of apelin-13 in angiogenesis in rat primary myocardial microvascular endothelial cells (MMVECs) and the mechanisms involved in it. In addition, compound C, an AMPK inhibitor, and LY294002, an Akt inhibitor were used to investigate whether AMPK and Akt signaling participate in the pro-angiogenic process induced by apelin-13.

Materials and methods

Materials

Apelin-13 was purchased from GL Biochem Shanghai (Shanghai, China). Dulbecco’s modified Eagle’s medium (DMEM) and fetal bovine serum (FBS) were purchased from Gibco-BRL (Carlsbad, CA, USA). Phospho-AMPK (Thr-172), pan-α-AMPK, phospho-Akt (Ser-473) and phospho-eNOS (Ser-1179) antibodies were purchased from Cell Signaling Technology (Beverly, MA, USA). Akt, eNOS and GAPDH antibodies were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA). Compound C was purchased from Toronto Research Chemicals Inc. (North York, ON, Canada) and LY294002 from Calbiochem (San Diego, CA, USA). Secondary antibodies were purchased from Beijing Biosynthesis Biotechnology Co., Ltd. (Beijing, China). All antibodies were polyclonal, and sourced from rabbit.

Isolation and identification of MMVECs in rats

Wistar rats (80–100 g) were used for the isolation of primary MMVECs. The animal protocol was approved by the Animal Care Committee of Shanghai Jiao Tong University (Shanghai, China). Briefly, rats were anesthetized with sodium pentobarbital (60 mg/kg) and heparinized by intraperitoneal injection of sodium heparin (500 U/0.1 kg). Following thoracotomy, the heart was rapidly dislodged and washed in phosphate-buffered saline (PBS). The atria, visible connective tissue, the right ventricle and the epicardial and endocardial surfaces of the left ventricle were carefully removed and the remaining myocardial tissue was washed in PBS with antibiotics (penicillin-streptomycin; 15140-122; Life Technologies, Carlsbad, CA, USA) several times prior to cutting into 1-mm3 sections without visible vessels. Myocardial tissues were seeded on culture plates pre-coated with rat tail tendon gelatin and incubated at 37°C in a humidified atmosphere of 5% CO2 for 30 min. The tissues were cultured in DMEM (4500 mg/l; D-glucose) supplemented with 20% FBS, 50 U/ml heparin, 100 U/ml penicillin and 100 μg/ml streptomycin. The tissue sections were discarded after the cells began to grow and the medium was changed at 70 h intervals. MMVECs were identified by their typical ‘cobblestone’ appearance and by positive CD31 and CD34 immunostaining. MMVECs at the second passage were used for experiments. The cells were allowed to grow to 80–90% confluence and were used for further experimental analysis.

Cell proliferation assay

The proliferation rate of MMVECs was determined by the MTT assay. The cells were trypsinized and resuspended in 10% DMEM. The cells were seeded at 2×103 cells per well in 96-well plates and cultured in a humidified 5% CO2 atmosphere at 37°C for 24 h. Drugs (apelin-13, compound C and LY294002) were added to the medium at the indicated times with different concentrations. Following incubation, the cells were washed with PBS and subsequently incubated with MTT at 37°C for 4 h. Following incubation, dimethylsulfoxide was added, the cells were incubated for 10 min and the absorbance at 490 nm was recorded with a Epoch Microplate spectrophotometer (BioTek Instruments, Inc., Winooski, VT, USA).

Migration assays
Cell scratch assay

MMVECs were trypsinized and resuspended in 10% DMEM. The cells were incubated in 6-well culture dishes in DMEM containing 10% FBS until 90% confluency. Following serum starvation for 24 h, a linear wound was made by scratching the bottom of the dish. Following wounding, cells were washed with PBS and incubated in serum-free DMEM. Following treatment, cells were allowed to migrate for 0, 8, 16 and 24 h. The wound width was measured in three areas selected at random and images were captured using the Motic AE31 Photometry and Dimensioning microscope (Milton, MA, USA). Migration was quantified by the assessment of the migration distance beyond the reference line using the Motic Image Plus software (Houston, TX, USA).

Boyden chamber assay

The Boyden chamber assay was performed as described previously (Neuroprobe, Cabin John, MD, USA) (18). Cells were trypsinized and resuspended in 10% DMEM. DMEM (800 μl/well; 10%) containing various concentrations of apelin-13, compound C and LY294002 was added into the wells in the lower chamber and 1.5×104 cells (200 μl/well) were added in the upper chamber. The chambers were incubated for 8–12 h at 37°C in a 5% CO2 humidified incubator. The cells migrating through the filter (cells on the lower side of the filter) were fixed in 4% paraformaldehyde for 10 min, stained with 0.1% crystal violet stain solution (Sigma, St. Louis, MO, USA) for 30 min and then five random microscopic fields per well were quantified. Each experiment was performed twice in triplicate.

Tube formation assay

The formation of vascular-like structures by MMVECs on growth factor-reduced Matrigel (BD Biosciences, Franklin Lakes, NJ, USA) was performed as previously described (19) and 24-well culture plates were coated with Matrigel according to the manufacturer’s instructions. The MMVECs were seeded on coated plates at 5×104 cells/well in DMEM containing 10% FBS and incubated at 37°C for 16–18 h. Tube formation was observed using an inverted phase contrast microscope (Nikon, Tokyo, Japan). Images were captured with a video graphic system (DEI-750 CE Digital Output Camera; Optronics, Goleta, CA, USA). The degree of tube formation was quantified by measuring the length of tubes from three randomly selected low power fields (x100) from each well using the NIH Image Program. Each experiment was repeated three times.

Western blot analysis

Western blot analysis was performed as previously described (20). In brief, cell lysates were extracted with Nonidet P-40 lysis buffer followed by SDS-PAGE. The membranes were immunoblotted with the indicated primary antibodies at a 1:1,000 dilution overnight, followed by the secondary antibody conjugated with horseradish peroxidase at a 1:2,000 dilution for 2 h. A western blotting detection kit (Amersham Biosciences, Piscataway, NJ, USA) and enhanced chemiluminescence reagent (Thermo Fisher Scientific, Waltham, MA, USA) were used to examine the membranes. Protein bands were evaluated by densitometry using ImageJ software (version 1.41; National Institutes of Health, Bethesda, MD, USA) and normalized to the expression levels of GAPDH for protein loading. Phospho-specific signals, normalized against the quantity of total protein, are shown as arbitrary units. The changes in phosphorylation were calculated by comparing the differences between basal and stimulated values of specifically treated samples and their respective controls.

Statistical analysis

The results are expressed as the mean ± standard deviation. Statistical comparisons were performed using analysis of variance with Scheffe’s F-test for post hoc analysis. P<0.05 was considered to indicate a statistically significant difference. Statistical analyses were performed using the SPSS 13.0 statistical software (SPSS, Chicago, IL, USA).

Results

Apelin-13 promotes the proliferation, migration and tube formation of MMVECs

The effect of apelin-13 on the proliferation of MMVECs was examined using the MTT assay. It was revealed that apelin-13 was able to promote the proliferation of MMVECs in a dose-dependent manner with a maximum effect being observed at 200 nmol/l (Fig. 1). The role of apelin-13 on endothelial cell migration was further evaluated by the scratch assay and the modified Boyden chamber assay. Apelin-13 significantly stimulated the migration of cells in a dose-dependent manner with an optimal activity at 200 nmol/l (Fig. 2A and B). To examine the effects of apelin-13 on the differentiation of MMVECs into vascular structures in vitro, tube formation ability was assessed. Apelin-13 significantly increased the tube formation of MMVECs in a dose-dependent manner with its optimal activity at 200 nmol/l (Fig. 2C).

Apelin-13 induces the phosphorylation of AMPK and Akt in MMVECs

It is known that AMPK and Akt are involved in the regulation of angiogenesis (11,15). The effect of apelin-13 on the phosphorylation status of AMPK at Thr-172 and Akt at Ser-473 was investigated in MMVECs. The treatment of MMVECs with apelin-13 enhanced the phosphorylation of AMPK in a dose- and time-dependent manner with maximal AMPK phosphorylation occurring at 200 nmol/l following 2 h exposure. Similarly, the phosphorylation of Akt was induced by apelin-13; however, this response was transient (Fig. 3A and C). Cross-talk between AMPK and Akt was previously described to regulate the phosphorylation of eNOS (11,21). Therefore, eNOS phosphorylation in MMVECs was also examined. Apelin-13 stimulated eNOS phosphorylation at Ser-1179 (p-eNOS) in a time- and dose-dependent manner, while the total protein levels (eNOS) remained unchanged. (Fig. 3A and C). Subsequently, MMVECs were treated with the AMPK and Akt inhibitors, it was revealed that they significantly suppressed the apelin-13-induced AMPK, Akt and eNOS phosphorylation in MMVECs (Fig. 3E and G), while incubation with the Akt inhibitor did not alter the endogenous phosphorylation of AMPK (Fig. 3G). Therefore, it was hypothesized that there is cross-talk between AMPK and Akt in the pro-angiogenic process induced by apelin-13.

AMPK and Akt signaling are required for apelin-13-stimulated migration and differentiation

To determine whether AMPK and Akt signaling are involved in apelin-13-stimulated endothelial cell migration and tube formation in vitro, MMVECs were treated either with compound C or LY294002, the inhibitors of AMPK and Akt, respectively. Cell scratch and Boyden chamber assays were used to assess the role of AMPK and Akt signaling on the migration ability of MMVECs induced by apelin-13. It was observed that the AMPK and Akt inhibitors significantly suppressed the apelin-13-stimulated migration of MMVECs (Fig. 4A and C). Subsequently, Matrigel assays were performed to investigate the effect of these two inhibitors on the tube formation induced by apelin-13 in MMVECs. The inhibitors significantly impaired the apelin-13-stimulated tube formation (Fig. 4E) of MMVECs. These data indicated that AMPK and Akt signaling are required for apelin-13-induced endothelial migration and differentiation in MMVECs.

Discussion

In the present study, it was demonstrated that apelin-13 promotes angiogenesis through the modulation of AMPK and Akt signaling in MMVECs. It was further investigated how AMPK and Akt signaling participates in the pro-angiogenic process and it was found that the activation of eNOS is involved in the two processes.

The regulatory role of apelin-13 in endothelial cells has previously been investigated. It was revealed that apelin is expressed in vascular endothelial cells and participates in regulating their proliferation and regenerative angiogenesis (22,23,24). Calcitonin gene-related peptide and vascular endothelial growth factor were reported to promote angiogenesis by promoting AMPK and Akt signaling activation, respectively (25,26). AMPK and Akt directly phosphorylate eNOS (17,27), which is important in angiogenesis. By contrast, apelin-13 can regulate glucose and lipid metabolism (12,13) via activating AMPK signaling. We hypothesized that apelin-13 may promote angiogenesis through the activation of AMPK and Akt signaling in MMVECs. The results demonstrated that apelin-13 can promote angiogenesis in MMVECs. Apelin-13 stimulated the phosphorylation of AMPK and eNOS in a time- and dose-dependent manner, while the phosphorylation of Akt increased in the early stages and decreased at the later stages. AMPK and Akt inhibitors suppressed the apelin-13-induced AMPK, Akt and eNOS phosphorylation and also inhibited apelin-13-stimulated cell migration and differentiation. Notably, although the AMPK inhibitor suppressed the endogenous Akt phosphorylation, the Akt inhibitor had no effect on the endogenous phosphorylation of AMPK in MMVECs. The present data suggest that apelin-13 exerts pro-angiogenic effects in MMVECs through the modulation of AMPK and Akt signaling and the activation of eNOS, which is in accordance with previous studies (28,29,30). A study by Nagata et al (11) demonstrated that AMPK did not exhibit an effect on endothelial cell migration, tube formation and NO production under normoxia, which suggests that further in vitro and in vivo studies are required to gain a greater understanding of the role of endothelial cell AMPK in angiogenesis.

Of note, in the present study, it was not determined how apelin-13 regulates AMPK and Akt signaling. In addition, although a subunit of AMPK was identified as the major target, further evaluation is required regarding the specific subunit (α1 or α2) involved. However, it was demonstrated that the activation of AMPK and Akt signaling was associated with angiogenesis induced by apelin-13 in MMVECs. Notably, the impaired angiogenesis was associated with the inhibition of the pathways.

The imbalance between angiogenesis and cardiac hypertrophy is important in the transition from adaptive hypertrophy to pathological cardiac remodeling in heart failure. Apelin-13 has been demonstrated to exert cardiovascular protective effects (31,32). The results of the present study demonstrated that apelin-13 is a pro-angiogenic factor, suggesting that the exogenous supplementation of apelin-13 may improve cardiac remodeling in patients with heart failure.

Acknowledgements

This study was supported by the Natural Science Foundation of Shanghai Science and Technology Commission (grant no. 10ZR1422900 to Professor Mingya Liu) and partly supported by the National Natural Science Foundation of China (grant no. 81070110 to Professor Meng Wei).

References

1 

Tatemoto K, Hosoya M, Habata Y, et al: Isolation and characterization of a novel endogenous peptide ligand for the human APJ receptor. Biochem Biophys Res Commun. 251:471–476. 1998. View Article : Google Scholar : PubMed/NCBI

2 

Kawamata Y, Habata Y, Fukusumi S, et al: Molecular properties of apelin: tissue distribution and receptor binding. Biochim Biophys Acta. 1538:162–171. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Kälin RE, Kretz MP, Meyer AM, et al: Paracrine and autocrine mechanisms of apelin signaling govern embryonic and tumor angiogenesis. Dev Biol. 305:599–614. 2007.PubMed/NCBI

4 

Hosoya M, Kawamata Y, Fukusumi S, et al: Molecular and functional characteristics of APJ. Tissue distribution of mRNA and interaction with the endogenous ligand apelin. J Biol Chem. 275:21061–21067. 2000. View Article : Google Scholar : PubMed/NCBI

5 

O’Carroll AM, Selby TL, Palkovits M and Lolait SJ: Distribution of mRNA encoding B78/apj, the rat homologue of the human APJ receptor, and its endogenous ligand apelin in brain and peripheral tissues. Biochim Biophys Acta. 1492:72–80. 2000.PubMed/NCBI

6 

Cheng X, Cheng XS and Pang CC: Venous dilator effect of apelin, an endogenous peptide ligand for the orphan APJ receptor, in conscious rats. Eur J Pharmacol. 470:171–175. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Szokodi I, Tavi P, Földes G, et al: Apelin, the novel endogenous ligand of the orphan receptor APJ, regulates cardiac contractility. Circ Res. 91:434–440. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Farkasfalvi K, Stagg MA, Coppen SR, et al: Direct effects of apelin on cardiomyocyte contractility and electrophysiology. Biochem Biophys Res Commun. 357:889–895. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Ribatti D, Vacca A, Roncali L and Dammacco F: The chick embryo chorioallantoic membrane as a model for in vivo research on angiogenesis. Int J Dev Biol. 40:1189–1197. 1996.PubMed/NCBI

10 

Mu J, Brozinick JT Jr, Valladares O, Bucan M and Birnbaum MJ: A role for AMP-activated protein kinase in contraction- and hypoxia-regulated glucose transport in skeletal muscle. Mol Cell. 7:1085–1094. 2001. View Article : Google Scholar : PubMed/NCBI

11 

Nagata D, Mogi M and Walsh K: AMP-activated protein kinase (AMPK) signaling in endothelial cells is essential for angiogenesis in response to hypoxic stress. J Biol Chem. 278:31000–31006. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Zhu S, Sun F, Li W, et al: Apelin stimulates glucose uptake through the PI3K/Akt pathway and improves insulin resistance in 3T3-L1 adipocytes. Mol Cell Biochem. 353:305–313. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Yue P, Jin H, Xu S, et al: Apelin decreases lipolysis via G(q), G(i), and AMPK-Dependent Mechanisms. Endocrinology. 152:59–68. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Chandra SM, Razavi H, Kim J, et al: Disruption of the apelin-APJ system worsens hypoxia-induced pulmonary hypertension. Arterioscler Thromb Vasc Biol. 31:814–820. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Shiojima I and Walsh K: Role of Akt signaling in vascular homeostasis and angiogenesis. Circ Res. 90:1243–1250. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Morales-Ruiz M, Fulton D, Sowa G, Languino LR, Fujio Y, Walsh K and Sessa WC: Vascular endothelial growth factor-stimulated actin reorganization and migration of endothelial cells is regulated via the serine/threonine kinase Akt. Circ Res. 86:892–896. 2000. View Article : Google Scholar

17 

Luo Z, Fujio Y, Kureishi Y, et al: Acute modulation of endothelial Akt/PKB activity alters nitric oxide-dependent vasomotor activity in vivo. J Clin Invest. 106:493–499. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Witzenbichler B, Kureishi Y, Luo Z, Le Roux A, Branellec D and Walsh K: Regulation of smooth muscle cell migration and integrin expression by the Gax transcription factor. J Clin Invest. 104:1469–1480. 1999. View Article : Google Scholar : PubMed/NCBI

19 

Kureishi Y, Luo Z, Shiojima I, et al: The HMG-CoA reductase inhibitor simvastatin activates the protein kinase Akt and promotes angiogenesis in normocholesterolemic animals. Nat Med. 6:1004–1010. 2000. View Article : Google Scholar : PubMed/NCBI

20 

Nagata D, Suzuki E, Nishimatsu H, et al: Cyclin A downregulation and p21(cip1) upregulation correlate with GATA-6-induced growth arrest in glomerular mesangial cells. Circ Res. 87:699–704. 2000. View Article : Google Scholar : PubMed/NCBI

21 

Kovacic S, Soltys CL, Barr AJ, Shiojima I, Walsh K and Dyck JR: Akt activity negatively regulates phosphorylation of AMP-activated protein kinase in the heart. J Biol Chem. 278:39422–39427. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Kleinz MJ and Davenport AP: Immunocytochemical localization of the endogenous vasoactive peptide apelin to human vascular and endocardial endothelial cells. Regul Pept. 118:119–125. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Eyries M, Siegfried G, Ciumas M, Montagne K, Agrapart M, Lebrin F and Soubrier F: Hypoxia-induced apelin expression regulates endothelial cell proliferation and regenerative angiogenesis. Circ Res. 103:432–440. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Masri B, Morin N, Cornu M, Knibiehler B and Audigier Y: Apelin (65–77) activates p70 S6 kinase and is mitogenic for umbilical endothelial cells. FASEB J. 18:1909–1911. 2004.

25 

Zheng S, Li W, Xu M, et al: Calcitonin gene-related peptide promotes angiogenesis via AMP-activated protein kinase. Am J Physiol Cell Physiol. 299:C1485–C1492. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Radisavljevic Z, Avraham H and Avraham S: Vascular endothelial growth factor up-regulates ICAM-1 expression via the phosphatidylinositol 3 OH-kinase/AKT/Nitric oxide pathway and modulates migration of brain microvascular endothelial cells. J Biol Chem. 275:20770–20774. 2000. View Article : Google Scholar

27 

Morrow VA, Foufelle F, Connell JM, Petrie JR, Gould GW and Salt IP: Direct activation of AMP-activated protein kinase stimulates nitric-oxide synthesis in human aortic endothelial cells. J Biol Chem. 278:31629–31639. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Zhu Z, Fu C, Li X, et al: Prostaglandin E2 promotes endothelial differentiation from bone marrow-derived cells through AMPK activation. PLoS One. 6:e235542011. View Article : Google Scholar : PubMed/NCBI

29 

Su KH, Yu YB, Hou HH, et al: AMP-activated protein kinase mediates erythropoietin-induced activation of endothelial nitric oxide synthase. J Cell Physiol. 227:3053–3062. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Izumi Y, Shiota M, Kusakabe H, et al: Pravastatin accelerates ischemia-induced angiogenesis through AMP-activated protein kinase. Hypertens Res. 32:675–679. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Tao J, Zhu W, Li Y, et al: Apelin-13 protects the heart against ischemia-reperfusion injury through inhibition of ER-dependent apoptotic pathways in a time-dependent fashion. Am J Physiol Heart Circ Physiol. 301:H1471–H1486. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Rastaldo R, Cappello S, Folino A, et al: Apelin-13 limits infarct size and improves cardiac postischemic mechanical recovery only if given after ischemia. Am J Physiol Heart Circ Physiol. 300:H2308–H2315. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2014
Volume 9 Issue 5

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
Yang X, Zhu W, Zhang P, Chen K, Zhao L, Li J, Wei M and Liu M: Apelin-13 stimulates angiogenesis by promoting cross‑talk between AMP-activated protein kinase and Akt signaling in myocardial microvascular endothelial cells. Mol Med Rep 9: 1590-1596, 2014
APA
Yang, X., Zhu, W., Zhang, P., Chen, K., Zhao, L., Li, J. ... Liu, M. (2014). Apelin-13 stimulates angiogenesis by promoting cross‑talk between AMP-activated protein kinase and Akt signaling in myocardial microvascular endothelial cells. Molecular Medicine Reports, 9, 1590-1596. https://doi.org/10.3892/mmr.2014.1984
MLA
Yang, X., Zhu, W., Zhang, P., Chen, K., Zhao, L., Li, J., Wei, M., Liu, M."Apelin-13 stimulates angiogenesis by promoting cross‑talk between AMP-activated protein kinase and Akt signaling in myocardial microvascular endothelial cells". Molecular Medicine Reports 9.5 (2014): 1590-1596.
Chicago
Yang, X., Zhu, W., Zhang, P., Chen, K., Zhao, L., Li, J., Wei, M., Liu, M."Apelin-13 stimulates angiogenesis by promoting cross‑talk between AMP-activated protein kinase and Akt signaling in myocardial microvascular endothelial cells". Molecular Medicine Reports 9, no. 5 (2014): 1590-1596. https://doi.org/10.3892/mmr.2014.1984