Open Access

Calycosin‑7‑O‑β‑D‑glucoside attenuates ischemia‑reperfusion injury in vivo via activation of the PI3K/Akt pathway

  • Authors:
    • Min Ren
    • Xudong Wang
    • Guoqing Du
    • Jiawei Tian
    • Yujie Liu
  • View Affiliations

  • Published online on: November 24, 2015     https://doi.org/10.3892/mmr.2015.4611
  • Pages: 633-640
  • Copyright: © Ren et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate the effects and mechanisms of calycosin‑7‑O‑β‑D‑glucoside (CG) on ischemia‑reperfusion (I/R) injury in vivo. Hemodynamic parameters, including ejection fraction (EF), fractional shortening (FS), left ventricular end‑systolic pressure (LVESP) and left ventricular end‑diastolic pressure (LVEDP) were monitored using an ultrasound system, and infarct size was measured using Evans blue/tetrazolium chloride double staining. The activities of serum creatine kinase (CK), lactate dehydrogenase (LDH) and superoxide dismutase (SOD), and the levels of malondialdehyde (MDA) were determined to assess the degree of myocardial injury and oxidative stress‑induced damage. The protein expression levels of cleaved‑caspase‑3, cleaved‑caspase‑9, phosphorylated (p)‑phosphatidylinositol 3‑kinase (PI3K) p85, PI3K p85, p‑Akt and Akt were determined using western blotting. The results demonstrated that pretreatment with high dose (H)‑CG markedly improved cardiac function, as evidenced by upregulated EF, FS and LVESP, and downregulated LVEDP. In addition, administration of CG resulted in significant decreases in infarct size in the I/R+low dose‑CG and I/R+H‑CG groups, compared with the I/R group. The activities of CK and LDH, and the levels of MDA in the I/R+H‑CG group were reduced, compared with those in the I/R group, whereas SOD activity was elevated. Treatment with CG inhibited the cleavage and activity of caspase‑3 and caspase‑9, and enhanced the phosphorylation of PI3K p85 and Akt. Notably, administration of the PI3K inhibitor, LY294002, markedly lowered the levels of p‑PI3K p85/p‑Akt, and eradicated the inhibitory effects of H‑CG on infarct size, myocardial injury and oxidative stress‑induced damage. Taken together, the results suggested that CG may alleviate I/R injury by activating the PI3K/Akt signaling pathway.

Introduction

Acute myocardial infarction (AMI) is associated with high mortality rates worldwide (1). AMI often occurs due to rupture of an atherosclerotic plaque in a coronary artery, which may induce thrombosis and artery occlusion, resulting in loss of blood supply to the affected area and necrosis. Annually, over 3,000,000 individuals suffer from acute ST-elevation myocardial infarction, and over 4,000,000 individuals suffer from non-ST-elevation myocardial infarction (2). Currently, the therapeutic strategies considered most effective are mechanical revascularization by percutaneous coronary intervention (3), thrombolytic therapy (4), primary angioplasty (5), coronary artery bypass grafting and antithrombotic therapy combined with timely reperfusion (6). However, these treatments are unable to improve cardiac function (7). In addition, tissue ischemia followed by reperfusion initiates systemic inflammation, which may aggravate local injury and induce remote multi-organ dysfunction (8). Therefore, the development of a safer, more effective strategy for reducing I/R injury and improving postoperative survival rates is required.

Astragali Radix (AR), the root of Astragalus membranaceus and Astragalus membranaceus var. mongholicus (9), is a traditional Chinese medicine (10). AR exerts various bioactivities, including antioxidation, enhancement of cardiovascular function, hepatoprotection, immunostimulation and myocardial protection in diabetic nephropathy (11). AR has also been reported to reduce myocardial ischemic injury (12), and AR extracts efficiently protect MRC-5 cells from H2O2-induced oxidative damage via the inhibition of superoxide dismutase (SOD) and catalase (13). A previous clinical report indicated that AR may be a promising agent in the treatment of acute cerebral infarction (14). AR contains various active components, including polysaccharides, flavonoids, astragalosides I–VII (saponins), amino acids and trace elements (15,16). Calycosin-7-O-β-d-glucoside (CG) is a predominant flavonoid of AR (1719), which is known to possess anti-inflammatory (20) and anti-osteoarthritic properties (21). A previous study has shown that CG significantly reduces cerebral infarct size and histological damage in a rat model of I/R. In addition, CG protects blood-brain barrier integrity by inhibiting the activities of matrix metalloproteinases, scavenging nitric oxide and promoting the expression of caveolin-1 (22). However, the effects of CG on myocardial I/R injury and the underlying mechanisms remain to be fully elucidated.

In the present study, a rat model of myocardial I/R injury was treated with CG, and the underlying molecular mechanisms of CG on myocardial I/R injury were evaluated.

Materials and methods

Animals

Male Wistar rats (8-week-old) were purchased from Charles River Laboratories (Beijing, China). Experiments were performed according to the guidelines for the animal care and use of laboratory animal protocols, and were approved by the Ethics Committee of The Second Affiliated Hospital of Harbin Medical University (Harbin, China; approval no. SCXK-2012-0001). The rats were maintained in an air-conditioned room with a constant temperature of 22°C and an alternating 12 h light/12 h dark cycle. The rats were provided with access to water and a standard diet ad libitum.

In vivo myocardial I/R model and experimental groups

The rats were anesthetized with 10% chloral hydrate (3 ml/kg body weight; Sinopharm Medicine, Shenyang, China) by intra-peritoneal injection. The rats were intubated, and mechanical ventilation was achieved by connecting the endotracheal tube to a scientific ventilator (HX-300S; Chengdu Technology & Market Co., Ltd, Chengdu, China) at a respiratory rate of 80 breaths/min with a tidal volume of 6–8 ml/kg body weight. A left thoracotomy was performed, in order to expose the heart and the root of the large blood vessel. The left anterior descending (LAD) coronary artery was subsequently transiently ligated with a nylon suture for a 45 min ischemic period. Microsurgical scissors were used to release the ligature, and the heart was reperfused for 3 h.

The rats were randomly divided into five groups (12 animals per group). In the sham group, the rats underwent the described anesthetic and surgical procedures without ligation of the LAD coronary artery; in the I/R group, the rats underwent myocardial ischemia for 45 min and reperfusion for 3 h by ligation of the LAD coronary artery; in the I/R+H-CG group, the rats were pretreated with a high dose of CG (H-CG; 30 mg/kg body weight; Dalian Meilun Biological Technology Co., Ltd., Dalian, China) via intravenous injection 30 min prior to ligation of the LAD coronary artery; in the I/R+L-CG group, the rats were pretreated with a low dose of CG (L-CG; 15 mg/kg body weight) via intravenous injection 30 min prior to ligation of the LAD coronary artery; in the sham+H-CG group, the rats were pretreated with H-CG via intravenous injection, and then underwent the described anesthetic and surgical procedures without ligation of the LAD coronary artery.

Detection of cardiac function

Following reperfusion, an ultrasound system (IE33; Philips GmbH, Herrsching, Germany) was used to collect hemodynamic parameters, including ejection fraction (EF), fractional shortening (FS), left ventricular end-systolic pressure (LVESP) and left ventricular end-diastolic pressure (LVEDP). Blood samples were obtained for biochemical investigations and the hearts were removed for Evans blue/tetrazolium chloride (TTC) staining and western blotting.

Tissue staining

The LAD coronary artery was religated following I/R and 2–3 ml 2% Evans blue solution (Wokai, Shanghai, China) was transcardially perfused. The rats were administered with KCl solution via a marginal ear vein, and the heart was stopped in diastole. The heart was subsequently removed, washed with saline, and maintained at −20°C for 30–60 min, prior to being divided into six 2-mm sections. The sections were stained with 1% TTC (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China) at 37°C and images were captured using a digital camera (D3000; Nikon, Tokyo, Japan). The area at risk (AAR; red staining) indicating the ischemic area, the infarct area (IA; white staining) and non-ischemic area (blue staining) were analyzed using an Image Analysis system (Image Pro Plus 6.0; Media Cybernetics, Inc., Rockville, MD, USA). Infarct size was defined as a percentage of IA to AAR (%).

Detection of creatine kinase (CK), lactate dehydrogenase (LDH), SOD and malondialdehyde (MDA)

Following reperfusion, blood samples (5–8 ml/mouse) were collected from the carotid artery and serum was obtained by centrifugation (1,111 × g, 10 min, 4°C). Commercial assay kits (Nanjing Jiancheng Bioengineering Institute, Nanjing, China) were used to detect the activities of CK (cat. no. A032), SOD (cat. no. A001-3), LDH (cat. no. A020-1) and MDA (cat. no. A003-1) in the serum, according to the manufacturer's protocol.

Western blot analysis

Total proteins were extracted from the AAR tissues using radioimmunoprecipitation assay lysis buffer (50 mM Tris, 150 mM NaCl, 1% Triton X-100, 1% sodium deoxycholate, 0.1% SDS, pH 7.4) (Beyotime Institute of Biotechnology, Haimen, China) and protein concentrations were determined using a Bichinchoninic Acid Protein Assay kit (Beyotime Institute of Biotechnology). Subsequently, 40 µg protein was separated by 10 or 13% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred onto polyvinylidene fluoride membranes (EMD Millipore, Bedford, MA, USA). The membranes were blocked with 5% nonfat milk or 1% bovine serum albumin (Amresco, Framingham, MA, USA), and then incubated with the following primary antibodies at 4°C overnight: Rabbit anti-rat cleaved-caspase-3 polyclonal antibody (pAb) (1:1,000 dilution; cat. no. WL0146); rabbit anti-rat cleaved-caspase-9 pAb (1:1,000 dilution; cat. no. WL0191); rabbit anti-rat phosphatidylinositol 3-kinase (PI3K) p85 pAb (1:1,000 dilution; cat. no. WL0191); rabbit anti-rat phosphorylated (p)-Akt pAb (1:1,000 dilution; cat. no. WLP001); rabbit anti-rat Akt pAb (1:1,000 dilution; cat. no. WL0003) (all Wanleibio, Shenyang, China) and rabbit anti-rat p-PI3K p85 pAb (1:500 dilution; cat. no. bs-5538R; Bioss, Beijing, China). The membranes were then washed with Tris-buffered saline containing Tween 20 (Beijing Solarbio Science & Technology Co., Ltd.), and incubated with horseradish peroxidase-conjugated goat anti-rabbit immunoglobulin G (1:5,000, Beyotime Institute of Biotechnology) at 37°C for 45 min. Band densities were analyzed using Gel-Pro Analyzer software 4.0 (Media Cybernetics, Inc.) and normalized to β-actin.

Detection of caspase-3/9 activity

Caspase-3/9 activity was measured using a Caspase Activity Assay kit (Beyotime Institute of Biotechnology), according to the manufacturer's protocol. Briefly, the total cellular proteins were quantified and reacted with the corresponding substrates: Ac-DEVD-ρNA or Ac-LEHD-ρNA. Caspase-3/9 activity was subsequently measured as the optical density of the cleaved substrate ρNA at 405 nm using a microplate reader (ELX-800; Bio-Tek Instruments, Inc., Winooski, VT, USA).

PI3K/Akt signaling pathway

The rats were randomly divided into three groups of 12: The I/R group, I/R+H-CG group and I/R+H-CG+LY294002 group. The PI3K inhibitor, LY294002, (0.3 mg/kg body weight; Sigma-Aldrich) was administered to the rats in the I/R+H-CG+LY294002 group via intravenous injection 30 min prior to the administration of H-CG. The rats were then subjected to I/R. Heart tissues from the AAR was lysed with lysis buffer and the expression levels of PI3K p85, p-PI3K p85, Akt and p-Akt were measured using western blot analysis. Infarct size was assessed using Evans blue/TTC double staining. The serum was obtained and levels of CK, SOD, LDH and MDA were analyzed using commercial kits (Nanjing Jiancheng Bioengineering Institute) as described above.

Statistical analysis

GraphPad Prism 5 software (GraphPad Software, Inc., La Jolla, CA, USA) was used for statistical analysis and image processing. Data are expressed as the mean ± standard deviation. Comparisons between the experimental groups were conducted using one-way analysis of variance, followed by a Bonferroni post-hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

CG ameliorates I/R-induced cardiac dysfunction

Ultrasound analysis was performed to detect cardiac function. As shown in Fig. 1, H-CG had no effect on EF (Fig. 1A), FS (Fig. 1B), LVEDP (Fig. 1C) or LVESP (Fig. 1D), in the sham+H-CG group, compared with the sham group. However, EF, FS and LVESP levels were markedly lower in the I/R group, compared with those in the sham group (P<0.01), whereas LVEDP was significantly higher, compared with the sham group (P<0.01). Following treatment with H-CG, EF, FS and LVESP (P<0.05) were significantly increased, whereas the LVEDP was decreased (P<0.01).

CG reduces myocardial infarct size

To determine whether CG affected myocardial infarct size, the rats were pretreated with L-CG or H-CG, and then subjected to I/R. As shown in Fig. 2, no ischemic and necrotic areas were detected in the sham or the sham+H-CG groups. I/R significantly increased the infarct size (29.98±5.28, vs. 0%; P<0.01). As expected, compared with the I/R group (29.98±5.28%), the infarct size was significantly smaller in the I/R+L-CG group (22.74±4.00; P<0.05) and the I/R+H-CG group (16.22±5.15%; P<0.01)

CG attenuates I/R-induced myocardial injury and oxidative stress-induced damage

The effects of CG were also evaluated on I/R-induced myocardial injury and damaged from oxidative stress. The activities of serum CK (Fig. 3A; P<0.01) and LDH (Fig. 3B; P<0.01) were markedly elevated in the I/R group, compared with those in the sham group. Following treatment with L-CG, only CK activity was inhibited (P<0.01); however, treatment with H-CG markedly inhibited the activities of the two markers (P<0.01). The activity of SOD, (Fig. 3C; P<0.01) was significantly lower in the I/R group, compared with the sham group. By contrast, MDA content (Fig. 3D; P<0.01) was significantly higher, compared with the sham group. Pretreatment with H-CG effectively increased the activity of SOD (P<0.01) and decreased levels of MDA (P<0.01).

CG reduces the I/R-induced increased expression levels and activities of pro-apoptotic factors

The results of the present study demonstrated that caspase cleavage (Fig. 4A; P<0.01), and the activities of caspase-3 (Fig. 4B; P<0.01) and caspase-9 (Fig. 4C; P<0.01) were enhanced in the I/R group, compared with those in the sham group. Treatment with L-CG and H-CG markedly downregulated the levels of cleaved-caspase-3 (P<0.01) and cleaved-caspase-9 (L-CG, P<0.05; H-CG, P<0.01). In addition, caspase activity was significantly inhibited following treatment with L-CG (caspase-3, P<0.01; caspase-9, P<0.05) or H-CG (P<0.01).

CG increases the phosphorylation of PI3K p85 and Akt

The protein expression levels of p-PI3K p85 (Fig. 5A; P<0.01) and p-Akt (Fig. 5B, P<0.01) were downregulated in the I/R group, compared with the sham group, and were upregulated in the I/R+L-CG group (p-PI3K p85, P<0.05; p-Akt, P>0.05) and I/R+H-CG group (P<0.01). No statistical differences were observed between the groups in the expression levels of total PI3K p85 or total Akt.

LY294002 inhibits H-CG-induced activation of the PI3K/Akt pathway

To confirm that CG attenuated I/R injury in vivo via activation of the PI3K/Akt pathway, the PI3K inhibitor, LY294002, was administered to the rats prior to H-CG. Treatment with H-CG significantly increased the phosphorylation of PI3K and Akt (Fig. 6A; P<0.01); however, suppressing PI3K activity with LY294002 effectively inhibited H-CG-induced PI3K/Akt phosphorylation (P<0.01). Treatment with H-CG significantly decreased infarct size, compared with the I/R group (15.67±3.28, vs. 35.46±5.33%, respectively; P<0.01), as shown in Fig. 6B, however, infarct size was significantly higher in the I/R+H-CG+LY294002 group, compared with that in the I/R+H-CG group (27.81±4.10, vs. 15.67±3.28%, respectively; P<0.01). Treatment with H-CG significantly decreased CK activity (Fig. 6C; P<0.01), LDH activity (P<0.01) and MDA content (P<0.01), and significantly increased SOD activity (P<0.01). However, co-treatment with LY294002 attenuated these effects (CK, P<0.01; LDH, P<0.05; MDA, P<0.05; SOD, P<0.05).

Discussion

AMI is a leading contributor to morbidity and mortality rates worldwide (23-25). Reperfusion improves clinical symptoms in patients with AMI (26); however, restoration of blood flow following ischemia may result in I/R injury (27,28), which is involved in the development of myocardial necrosis, arrhythmia, myocardial stunning, endothelial dysfunction and microvascular complications (26,29). The present study demonstrated that CG may exert a cardioprotective effect in a rat model of I/R-induced injury via the PI3K/Akt signaling pathway.

Previous studies have indicated that myocardial I/R injury alters hemodynamic parameters and affects cardiac function (30,31). In addition, levels of EF, FS (32,33) and LVSP are lower in I/R groups than in sham groups, whereas, LVEDP levels are higher (34,35), which is in agreement with the results of the present study. The present study also demonstrated that treatment with H-CG significantly restored the I/R-induced downregulation of EF, FS and LVESP, and markedly lowered the levels of LVEDP in the I/R+H-CG group, compared with the I/R group. These results suggested that CG improved cardiac function in the rat model of I/R.

Myocardial infarct size is an indicator of myocardial injury, and I/R has been reported to result in infarction in MI/R groups, compared with sham groups (36,37). Treatment with CG has been shown to significantly reduce infarct volume in a rat model of middle cerebral artery occlusion cerebral I/R injury (22). Consistently, the present study observed that L-CG and H-CG efficiently decreased infarct size.

LDH (38) and CK (39) are often elevated in MI, and are used to assess the degree of myocardial injury. Numerous evidence has demonstrated that I/R often induces the generation of reactive oxygen species (ROS) and oxidative stress (4042). Subsequently, ROS interacts with cell membrane lipids and produces MDA, which impairs cardiac function and induces myocardial cell injury (43,44). Therefore, reducing oxidative stress is an advantageous strategy for the alleviation of I/R injury. In the present study, CK and LDH activity, and MDA content were increased in the I/R group, compared with in the sham group; however, SOD activity was decreased, which was consistent with the results of previous studies (37,45). These results indicated that CG exerted its cardioprotective effects by notably decreasing CK, LDH and MDA, and increasing SOD activity.

Apoptosis is important in development and tissue homeostasis (46), and caspases are considered the executioners of apoptosis (47). Once cells receive apoptotic stimuli, the mitochondrial outer membrane becomes permeabilized and cytochrome c is released from the mitochondria into the cytosol (48). Cytochrome c interacts with apoptotic protease activating factor 1 and procaspase-9, which is cleaved into caspase-9 and initiates the activation of caspase-3, caspase-6 and caspase-7 (49). Previous studies have reported that I/R injury is associated with the apoptosis of cardiomyocytes (50,51). The present study demonstrated that the expression levels of cleaved-caspase-3 and cleaved-caspase-9, and caspase activity were downregulated in the I/R+L-CG and I/R+H-CG groups, compared with in the I/R group. Therefore, CG may alleviate I/R injury by suppressing caspase activity and inhibiting cardiomyocyte apoptosis.

PI3K consists of a catalytic subunit (p110) and a regulatory subunit (p85) (52,53). Akt is a serine-threonine kinase and, following phosphorylation, performs its antiapoptotic effects via the activation of B-cell lymphoma-2-associated death promoter and caspases (54). Previous studies have reported that the PI3K/Akt signaling pathway is crucial in protecting the myocardium from MI/R injury (55), and the activation of PI3K/Akt significantly reduces cardiomyocyte apoptosis (56). The present study examined the expression levels of PI3K p85, p-PI3K p85, Akt and p-Akt. Pretreatment with CG effectively activated and phosphorylated PI3K and Akt, whereas the levels of total PI3K p85 and Akt were not changed. The PI3K inhibitor, LY294002, was used to determine whether the PI3K/Akt pathway was involved in the CG-mediated alleviation of I/R injury. Suppressing PI3K activity with LY294002 reversed the beneficial effects of CG. Based on the above results, it was hypothesized that CG alleviates I/R injury by activating the PI3K/Akt signaling pathway.

In conclusion, the results of the present study demonstrated that CG attenuated myocardial I/R injury in the rat model. The protective effects may be associated with activation of the PI3K/Akt pathway, and the inhibition of oxidative stress and pro-apoptotic factors.

Acknowledgments

The present study was supported by grants from the National Natural Science Foundation of China (grant nos. 81171404 and 81201093).

References

1 

Murray CJ and Lopez AD: Mortality by cause for eight regions of the world: Global Burden of Disease Study. Lancet. 349:1269–1276. 1997. View Article : Google Scholar : PubMed/NCBI

2 

White HD and Chew DP: Acute myocardial infarction. Lancet. 372:570–584. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Betgem RP, de Waard GA, Nijveldt R, Beek AM, Escaned J and van Royen N: Intramyocardial haemorrhage after acute myocardial infarction. Nat Rev Cardiol. 12:156–167. 2015. View Article : Google Scholar

4 

Lincoff AM and Topol EJ: Illusion of reperfusion. Does anyone achieve optimal reperfusion during acute myocardial infarction? Circulation. 88:1361–1374. 1993. View Article : Google Scholar : PubMed/NCBI

5 

Hartwell D, Colquitt J, Loveman E, Clegg AJ, Brodin H, Waugh N, Royle P, Davidson P, Vale L and MacKenzie L: Clinical effectiveness and cost-effectiveness of immediate angioplasty for acute myocardial infarction: Systematic review and economic evaluation. Health Technol Assess. 9:1–99. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Luo J, Xu H and Chen KJ: Potential benefits of Chinese Herbal Medicine for elderly patients with cardiovascular diseases. J Geriatr Cardiol. 10:305–309. 2013.

7 

Clifford DM, Fisher SA, Brunskill SJ, Doree C, Mathur A, Watt S and Martin-Rendon E: Stem cell treatment for acute myocardial infarction. Cochrane Database Syst Rev. 2:CD0065362012.PubMed/NCBI

8 

Abela CB and Homer-Vanniasinkham S: Clinical implications of ischaemia-reperfusion injury. Pathophysiology. 9:229–240. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Yu KZ, Liu J, Guo BL, Zhao ZZ, Hong H, Chen HB and Cai SQ: Microscopic research on a multi-source traditional Chinese medicine, Astragali Radix. J Nat Med. 68:340–350. 2014. View Article : Google Scholar

10 

Ismail ZM, Amin NM, Yacoub MF and Mohamed AM: Myelo-enhancement by astragalus membranaceus in male albino rats with chemotherapy myelo-suppression. Histological and immunohistochemical study. Int J Stem Cells. 7:12–22. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Liu XB, Ma L, Zhang AH, Zhang YH, Jiang J, Ma W, Zhang LM, Ren WC and Kong XJ: High-throughput analysis and characterization of Astragalus membranaceus transcriptome using 454 GS FLX. PLoS One. 9:e958312014. View Article : Google Scholar : PubMed/NCBI

12 

Jin Y, Chen Q, Li X, Fan X and Li Z: Astragali Radix protects myocardium from ischemia injury by modulating energy metabolism. Int J Cardiol. 176:1312–1315. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Xu X, Li F, Zhang X, Li P, Zhang X, Wu Z and Li D: In vitro synergistic antioxidant activity and identification of antioxidant components from Astragalus membranaceus and Paeonia lactiflora. PLoS One. 9:e967802014. View Article : Google Scholar

14 

Luo Y, Qin Z, Hong Z, Zhang X, Ding D, Fu JH, Zhang WD and Chen J: Astragaloside IV protects against ischemic brain injury in a murine model of transient focal ischemia. Neurosci Lett. 363:218–223. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Ko JK and Chik CW: The protective action of radix Astragalus membranaceus against hapten-induced colitis through modulation of cytokines. Cytokine. 47:85–90. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Ma XQ, Shi Q, Duan JA, Dong TT and Tsim KW: Chemical analysis of Radix Astragali (Huangqi) in China: A comparison with its adulterants and seasonal variations. J Agric Food Chem. 50:4861–4866. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Yu H, Zhang WL, Ding X, Zheng KY, Ho CM, Tsim KW and Lee YK: Optimizing combinations of flavonoids deriving from astragali radix in activating the regulatory element of erythropoietin by a feedback system control scheme. Evid Based Complement Alternat Med. 2013:5414362013. View Article : Google Scholar : PubMed/NCBI

18 

Zheng KY, Choi RC, Cheung AW, Guo AJ, Bi CW, Zhu KY, Fu Q, Du Y, Zhang WL, Zhan JY, et al: Flavonoids from Radix Astragali induce the expression of erythropoietin in cultured cells: A signaling mediated via the accumulation of hypoxia-inducible factor-1α. J Agric Food Chem. 59:1697–1704. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Lin LZ, He XG, Lindenmaier M, Nolan G, Yang J, Cleary M, Qiu SX and Cordell GA: Liquid chromatography-electrospray ionization mass spectrometry study of the flavonoids of the roots of Astragalus mongholicus and A. membranaceus. J Chromatogr A. 876:87–95. 2000. View Article : Google Scholar : PubMed/NCBI

20 

Li W, Sun YN, Yan XT, Yang SY, Kim S, Lee YM, Koh YS and Kim YH: Flavonoids from Astragalus membranaceus and their inhibitory effects on LPS-stimulated pro-inflammatory cytokine production in bone marrow-derived dendritic cells. Arch Pharm Res. 37:186–192. 2014. View Article : Google Scholar

21 

Pan H, Wang Y, Zhang Y, Zhou T, Fang C, Nan P, Wang X, Li X, Wei Y and Chen J: Phenylalanine ammonia lyase functions as a switch directly controlling the accumulation of calycosin and calycosin-7-O-beta-D-glucoside in Astragalus membranaceus var. mongholicus plants. J Exp Bot. 59:3027–3037. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Fu S, Gu Y, Jiang JQ, Chen X, Xu M, Chen X and Shen J: Calycosin-7-O-β-D-glucoside regulates nitric oxide/caveolin-1/matrix metalloproteinases pathway and protects blood-brain barrier integrity in experimental cerebral ischemia-reperfusion injury. J Ethnopharmacol. 155:692–701. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Ozaki K, Inoue K, Sato H, Iida A, Ohnishi Y, Sekine A, Sato H, Odashiro K, Nobuyoshi M, Hori M, et al: Functional variation in LGALS2 confers risk of myocardial infarction and regulates lymphotoxin-alpha secretion in vitro. Nature. 429:72–75. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Timmers L, Sluijter JP, van Keulen JK, Hoefer IE, Nederhoff MG, Goumans MJ, Doevendans PA, van Echteld CJ, Joles JA, Quax PH, et al: Toll-like receptor 4 mediates maladaptive left ventricular remodeling and impairs cardiac function after myocardial infarction. Circ Res. 102:257–264. 2008. View Article : Google Scholar

25 

Zou Y, Takano H, Mizukami M, Akazawa H, Qin Y, Toko H, Sakamoto M, Minamino T, Nagai T and Komuro I: Leukemia inhibitory factor enhances survival of cardiomyocytes and induces regeneration of myocardium after myocardial infarction. Circulation. 108:748–753. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Moens AL, Claeys MJ, Timmermans JP and Vrints CJ: Myocardial ischemia/reperfusion-injury, a clinical view on a complex pathophysiological process. Int J Cardiol. 100:179–190. 2005. View Article : Google Scholar : PubMed/NCBI

27 

Prasad A, Stone GW, Holmes DR and Gersh B: Reperfusion injury, microvascular dysfunction and cardioprotection: The 'dark side' of reperfusion. Circulation. 120:2105–2112. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Ahmed LA, Salem HA, Attia AS and El-Sayed ME: Enhancement of amlodipine cardioprotection by quercetin in ischaemia/reperfusion injury in rats. J Pharm Pharmacol. 61:1233–1241. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Buja LM and Weerasinghe P: Unresolved issues in myocardial reperfusion injury. Cardiovasc Pathol. 19:29–35. 2010. View Article : Google Scholar

30 

Ali N, Rizwi F, Iqbal A and Rashid A: Induced remote ischemic pre-conditioning on ischemia-reperfusion injury in patients undergoing coronary artery bypass. J Coll Physicians Surg Pak. 20:427–431. 2010.PubMed/NCBI

31 

Toldo S, Seropian IM, Mezzaroma E, Van Tassell BW, Salloum FN, Lewis EC, Voelkel N, Dinarello CA and Abbate A: Alpha-1 antitrypsin inhibits caspase-1 and protects from acute myocardial ischemia-reperfusion injury. J Mol Cell Cardiol. 51:244–251. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Fan Q, Chen M, Zuo L, Shang X, Huang MZ, Ciccarelli M, Raake P, Brinks H, Chuprun KJ, Dorn GW II, et al: Myocardial ablation of G protein-coupled receptor kinase 2 (GRK2) decreases ischemia/reperfusion injury through an anti-intrinsic apoptotic pathway. PLoS One. 8:e662342013. View Article : Google Scholar : PubMed/NCBI

33 

Zhou YC, Liu B, Li YJ, Jing LL, Wen G, Tang J, Xu X, Lv ZP and Sun XG: Effects of buyang huanwu decoction on ventricular remodeling and differential protein profile in a rat model of myocardial infarction. Evid Based Complement Alternat Med. 2012:3852472012. View Article : Google Scholar : PubMed/NCBI

34 

Song CL, Liu B, Diao HY, Shi YF, Li YX, Zhang JC, Lu Y, Wang G, Liu J, Yu YP, et al: The protective effect of microRNA-320 on left ventricular remodeling after myocardial ischemia-reperfusion injury in the rat model. Int J Mol Sci. 15:17442–17456. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Zhao G, Wang S, Wang Z, Sun A, Yang X, Qiu Z, Wu C, Zhang W, Li H, Zhang Y, et al: CXCR6 deficiency ameliorated myocardial ischemia/reperfusion injury by inhibiting infiltration of monocytes and IFN-γ-dependent autophagy. Int J Cardiol. 168:853–862. 2013. View Article : Google Scholar

36 

Wang Y, Li X, Wang X, Lau W, Wang Y, Xing Y, Zhang X, Ma X and Gao F: Ginsenoside Rd attenuates myocardial ischemia/reperfusion injury via Akt/GSK-3β signaling and inhibition of the mitochondria-dependent apoptotic pathway. PLoS One. 8:e709562013. View Article : Google Scholar

37 

Han J, Wang D, Yu B, Wang Y, Ren H, Zhang B, Wang Y and Zheng Q: Cardioprotection against ischemia/reperfusion by licochalcone B in isolated rat hearts. Oxid Med Cell Longev. 2014:1348622014. View Article : Google Scholar : PubMed/NCBI

38 

Kemp M, Donovan J, Higham H and Hooper J: Biochemical markers of myocardial injury. Br J Anaesth. 93:63–73. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Singh G, Rohilla A, Singh M and Balakumar P: Possible role of JAK-2 in attenuated cardioprotective effect of ischemic preconditioning in hyperhomocysteinemic rat hearts. Yakugaku Zasshi. 129:523–535. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Petrosillo G, Ruggiero FM, Di Venosa N and Paradies G: Decreased complex III activity in mitochondria isolated from rat heart subjected to ischemia and reperfusion: Role of reactive oxygen species and cardiolipin. FASEB J. 17:714–716. 2003.PubMed/NCBI

41 

Shimouchi A, Yokota H, Ono S, et al: Neuroprotective effect of water-dispersible hesperetin in retinal ischemia reperfusion injury. Jpn J Ophthalmol. Sept 25–2015.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

42 

Wang AL, Niu Q, Shi N, et al: Glutamine ameliorates intestinal ischemia-reperfusion Injury in rats by activating the Nrf2/Are signaling pathway. Int J Clin Exp Pathol. 8:7896–7904. 2015.PubMed/NCBI

43 

Kalaycioglu S, Sinci V, Imren Y and Oz E: Metoprolol prevents ischemia-reperfusion injury by reducing lipid peroxidation. Jpn Circ J. 63:718–721. 1999. View Article : Google Scholar : PubMed/NCBI

44 

Dhalla NS, Elmoselhi AB, Hata T and Makino N: Status of myocardial antioxidants in ischemia-reperfusion injury. Cardiovasc Res. 47:446–456. 2000. View Article : Google Scholar : PubMed/NCBI

45 

Zhang S, Li H and Yang SJ: Tribulosin protects rat hearts from ischemia/reperfusion injury. Acta Pharmacol Sin. 31:671–678. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Nakagawa T, Zhu H, Morishima N, Li E, Xu J, Yankner BA and Yuan J: Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-beta. Nature. 403:98–103. 2000. View Article : Google Scholar : PubMed/NCBI

47 

Riedl SJ and Shi Y: Molecular mechanisms of caspase regulation during apoptosis. Nat Rev Mol Cell Biol. 5:897–907. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Waterhouse NJ, Goldstein JC, von Ahsen O, Schuler M, Newmeyer DD and Green DR: Cytochrome c maintains mitochondrial transmembrane potential and ATP generation after outer mitochondrial membrane permeabilization during the apoptotic process. J Cell Biol. 153:319–328. 2001. View Article : Google Scholar : PubMed/NCBI

49 

Denault JB, Eckelman BP, Shin H, Pop C and Salvesen GS: Caspase 3 attenuates XIAP (X-linked inhibitor of apoptosis protein)-mediated inhibition of caspase 9. Biochem J. 405:11–19. 2007. View Article : Google Scholar : PubMed/NCBI

50 

Hamacher-Brady A, Brady NR, Logue SE, Sayen MR, Jinno M, Kirshenbaum LA, Gottlieb RA and Gustafsson AB: Response to myocardial ischemia/reperfusion injury involves Bnip3 and autophagy. Cell Death Differ. 14:146–157. 2007. View Article : Google Scholar

51 

Zhang W, Xing B, Yang L, Shi J and Zhou X: Icaritin attenuates myocardial ischemia and reperfusion injury via anti-inflammatory and anti-oxidative stress effects in rats. Am J Chin Med. 43:1083–1097. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Shin YK, Liu Q, Tikoo SK, Babiuk LA and Zhou Y: Influenza A virus NS1 protein activates the phosphatidylinositol 3-kinase (PI3K)/Akt pathway by direct interaction with the p85 subunit of PI3K. J Gen Virol. 88(Pt 1): 13–18. 2007. View Article : Google Scholar

53 

Ray PD, Huang BW and Tsuji Y: Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal. 24:981–990. 2012. View Article : Google Scholar : PubMed/NCBI

54 

Massion PP, Taflan PM, Shyr Y, Rahman SM, Yildiz P, Shakthour B, Edgerton ME, Ninan M, Andersen JJ and Gonzalez AL: Early involvement of the phosphatidylinositol 3-kinase/Akt pathway in lung cancer progression. Am J Respir Crit Care Med. 170:1088–1094. 2004. View Article : Google Scholar : PubMed/NCBI

55 

Zhang Y, Wei L, Sun D, Cao F, Gao H, Zhao L, Du J, Li Y and Wang H: Tanshinone IIA pretreatment protects myocardium against ischaemia/reperfusion injury through the phosphatidylinositol 3-kinase/Akt-dependent pathway in diabetic rats. Diabetes Obes Metab. 12:316–322. 2010. View Article : Google Scholar : PubMed/NCBI

56 

Fujio Y, Nguyen T, Wencker D, Kitsis RN and Walsh K: Akt promotes survival of cardiomyocytes in vitro and protects against ischemia-reperfusion injury in mouse heart. Circulation. 101:660–667. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2016
Volume 13 Issue 1

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
Ren M, Wang X, Du G, Tian J and Liu Y: Calycosin‑7‑O‑β‑D‑glucoside attenuates ischemia‑reperfusion injury in vivo via activation of the PI3K/Akt pathway. Mol Med Rep 13: 633-640, 2016
APA
Ren, M., Wang, X., Du, G., Tian, J., & Liu, Y. (2016). Calycosin‑7‑O‑β‑D‑glucoside attenuates ischemia‑reperfusion injury in vivo via activation of the PI3K/Akt pathway. Molecular Medicine Reports, 13, 633-640. https://doi.org/10.3892/mmr.2015.4611
MLA
Ren, M., Wang, X., Du, G., Tian, J., Liu, Y."Calycosin‑7‑O‑β‑D‑glucoside attenuates ischemia‑reperfusion injury in vivo via activation of the PI3K/Akt pathway". Molecular Medicine Reports 13.1 (2016): 633-640.
Chicago
Ren, M., Wang, X., Du, G., Tian, J., Liu, Y."Calycosin‑7‑O‑β‑D‑glucoside attenuates ischemia‑reperfusion injury in vivo via activation of the PI3K/Akt pathway". Molecular Medicine Reports 13, no. 1 (2016): 633-640. https://doi.org/10.3892/mmr.2015.4611