Open Access

Ononin alleviates H2O2‑induced cardiomyocyte apoptosis and improves cardiac function by activating the AMPK/mTOR/autophagy pathway

  • Authors:
    • Rongrong Pan
    • Qin Zhuang
    • Jiangtin Wang
  • View Affiliations

  • Published online on: September 16, 2021     https://doi.org/10.3892/etm.2021.10742
  • Article Number: 1307
  • Copyright: © Pan 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

Ononin (ON) is an isoflavone with numerous reported bioactivities, including anti‑oxidative, anti‑inflammatory and neuroprotective effects. Autophagy is a critical homeostatic process in the body that has been reported to closely associate with the apoptotic processes of cardiomyocytes. Using flow cytometry, western blotting, echocardiography and Masson's staining, the present study investigated the effects of ON on H2O2‑induced cardiomyocyte apoptosis and myocardial infarction, in addition to any potential underlying molecular mechanisms. H2O2 treatment reliably induced apoptosis in H9C2 cells. The anti‑apoptotic effects of ON were revealed by flow cytometry results and by the downregulation of cleaved‑caspase 3. Further investigations indicated that ON may alleviate apoptosis by enhancing autophagy, as evidenced by increased microtubule‑associated proteins 1A/1B light chain 3B expression and p62 degradation. Activation of the 5' AMP‑activated protein kinase (AMPK)/mTOR pathway was observed after ON administration following H2O2‑induced cardiomyocyte injury. However, these anti‑apoptotic effects mediated by ON were lost after autophagy inhibition by chloroquine or AMPK inhibition by Compound C. Finally, the protective effects of ON on cardiomyocytes in vitro could also be observed in vivo. A myocardial infarction model was established by ligating the left anterior descending branch of the rat heart. Using echocardiography and Masson's staining, ON was shown to increase the ejection fraction and decrease cardiac fibrosis in rats with myocardial infarction. These results suggest that ON exerts cardioprotective effects by improving autophagy via the AMPK/mTOR signaling pathway.

Introduction

Cardiovascular disease is one of the leading causes of human mortality worldwide, which is an important social and economic burden (1). The loss of cardiomyocytes during various ischemic heart diseases is considered to be irreversible, thereby contributing to permanent cardiac dysfunction (2). Apoptosis is a type of cell death that is characterized by prominent changes in caspase activation and serves an important role in cardiac injury (3). Therefore, the amelioration of apoptosis and improving cardiomyocyte viability are of particular research interest.

Autophagy is the phagocytosis and subsequent degradation of intracellular proteins or organelles for metabolism (4). Autophagy has been implicated in numerous physiological and pathological processes, including survival, differentiation, cancer, aging and heart disease (5). Emerging evidence has indicated that autophagy exerts a biphasic role in cardiac physiology, whereby excessive or reduced autophagy levels can both result in cardiomyocyte apoptosis (6,7). However, the exact mechanism underlying this remains unclear. mTOR is considered to be a key signaling component among the several proteins that have been reported to regulate autophagy (8). mTOR can directly or indirectly integrate cell signals, such as ATP and hypoxia, to regulate autophagy induction (9,10). In addition, as an important regulatory enzyme of metabolism, increased phosphorylation of 5'-AMP-activated protein kinase (AMPK) was previously found to inhibit autophagy (11).

Isoflavones are particularly abundant in legumes (such as soy, chickpea and red clover) and can also be found in various traditional Chinese medicines (such as Astragali Radix and Puerariae Lobatae Radix) (12,13). Previous studies have reported the protective effects of isoflavone in neuron injury, where it conferred antioxidant and anti-inflammatory properties (14,15). Ononin (ON) is a bioactive isoflavone of legumes, however, to the best of our knowledge, the specific functions and mechanisms of ON in cardiomyocytes have not been previously reported. Therefore, the present study aimed to assess the potential role of ON in H2O2-induced H9C2 cell injury and investigate how it could modulate the AMPK/mTOR/autophagy signaling pathway. Additionally, ON was studied in a rat myocardial infarction model to study its possible role in cardiac function.

Materials and methods

Drugs

ON (MedChemExpress), chloroquine (CQ; MedChemExpress) and Compound C (CC; Selleck Chemicals) were dissolved in DMSO (Sigma-Aldrich; Merck KGaA) and diluted with PBS.

Cell culture and treatment

The rat cardiomyocyte H9C2 cell line was purchased from the American Type Culture Collection. Cells were cultured in high-glucose DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (Beyotime Institute of Biotechnology) and 1% penicillin/streptomycin (Sigma-Aldrich; Merck KGaA) in an atmosphere containing 5% CO2 at 37˚C. The cells were subsequently randomly divided into the following treatment groups: i) Control group; ii) H2O2 group, which involved culturing in serum-free DMEM and exposed to H2O2 (400 µmol/l) to stimulate cardiomyocyte injury for 4 h at 37˚C; iii) H2O2 + ON group, which was treated with H2O2 and ON (10 µmol/l) simultaneously for 4 h at 37˚C; iv) H2O2 + ON + CC group, which was treated with H2O2, ON and CC (10 µM) simultaneously for 4 h at 37˚C; and (v) H2O2 + ON + CQ group, which was treated with H2O2, ON and CQ (10 µM) simultaneously for 4 h at 37˚C.

Cell viability assay

A Cell Counting Kit-8 (CCK-8; Dojindo Molecular Technologies, Inc.) was used to evaluate cell viability. A total of 1x104 H9C2 cells were plated onto 96-well plates and after treatment with H2O2 (400 µmol/l), 10 µl CCK-8 solution was added to each well and incubated at 37˚C for 2 h. Finally, the optical density absorbance of each well was measured at 450 nm using a microplate reader (Bio-Rad Laboratories, Inc.) to calculate the relative viability.

Flow cytometry assay

The FITC Annexin V Apoptosis Detection Kit (BD Biosciences) was used to evaluate H9C2 cell apoptosis, and the experimental procedures were conducted according to the manufacturer's protocols. After H2O2 treatment (400 µmol/l), H9C2 cells were collected, washed twice with PBS and transferred into a culture tube (5x104 cells/tube)before 5 µl Annexin V-FITC and 5 µl PI were added to the cells and incubated for 15 min at room temperature in the dark. BD FACSCalibur™ flow cytometer (BD Biosciences) and Cell Quest software version 3.1 were used to perform the subsequent apoptosis analysis.

Western blot analysis

Total protein from H9C2 cells or rat heart tissue was lysed using RIPA buffer (Cell Signaling Technology, Inc.) supplemented with protease and phosphatase inhibitors (Cell Signaling Technology, Inc.). Protein concentration was determined using a BCA kit (Beyotime Institute of Biotechnology). Protein lysate samples of 30 µg were separated by 10% SDS-PAGE and transferred onto PVDF membranes (EMD Millipore). The membranes were blocked with 8% non-fat milk at room temperature for 60 min, before being incubated with the following primary antibodies overnight at 4˚C: Cleaved-caspase-3 (1:500; Cell Signaling Technology, Inc., cat. no. 9664), caspase 3 (1:1,000; Cell Signaling Technology, Inc., cat. no. 9662), Bcl2 (1:800; Cell Signaling Technology, Inc., cat. no. 3869), Bax (1:1,000; Cell Signaling Technology, Inc., cat. no. 2772), GAPDH (1:10,000, Abcam, cat. no. ab9385), p62 (1:1,000; Cell Signaling Technology, Inc., cat. no. 5114), LC3-I (1:1,000; Cell Signaling Technology, Inc., cat. no. 4108), LC3-II (1:1,000; Cell Signaling Technology, Inc., cat. no. 4108), actin (1:10,000; Abcam, cat. no. ab20272), phosphorylated (p-) AMPK (1:1,000; Cell Signaling Technology, Inc., cat. no. 4186), AMPK (1:1,000; Cell Signaling Technology, Inc., cat. no. 5832), p-mTOR (1:1,000; Cell Signaling Technology, Inc., cat. no. 5536) and mTOR (1:1,000; Cell Signaling Technology, Inc.; cat. no. 2972). After washing with PBS with 0.1% Tween®-20 three times at 10 min each, the membranes were incubated with the corresponding HRP-conjugated secondary antibodies of goat anti-rabbit IgG (1:3,000; Cell Signaling Technology, Inc., no. 7074) and goat anti-mouse IgG (1:3,000; Cell Signaling Technology, Inc., no. 56970) for 1 h at room temperature. Finally, the blots were visualized using an ECL kit (EMD Millipore) and quantified using the Image Lab software version 4.0 (Bio-Rad Laboratories, Inc.) with GAPDH used as the loading control of Cleaved-caspase-3 and actin used as the loading control of the other proteins.

Myocardial infarction model and ononin treatment

All animal procedures were conducted according to the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health and were approved by the Animal Care and Use Committee of Wenzhou Medical University (Cixi, China). Male Sprague-Dawley rats (250-300 g, 6-8 weeks) were obtained from Experimental Animal Center of Wenzhou Medical University. Rats were maintained in climate- and light-controlled room (24±1˚C, relative humidity of 65±15%, 12/12-h light/dark cycle) with ad libitum access to food and water for at least 1 week prior to the experiments. Myocardial infarction (MI) was surgically induced as previously described (16). A total of 18 rats were anesthetized via an intraperitoneal injection of pentobarbital sodium (50 mg/kg) and ventilated with a rodent ventilator. Rats were divided into three groups (n=6/group): i) Sham; ii) MI and iii) MI + ON. The heart was then exposed, and the left anterior descending coronary artery was ligated permanently with a 6-0 silk suture in the MI group. The heart was exposed but the left anterior descending coronary artery was not ligated in the sham group. Rats in the treatment groups were intragastric injection of ON (20 mg/kg) (13) daily, whilst the MI group received the same volume of laboratory animal drinking water daily until 28 days after surgery.

Cardiac function assessment

Heart function was assessed using transthoracic echocardiography 28 days after MI induction. Left ventricular ejection fraction (LVEF) and left ventricular fraction shortening (LVFS) were analyzed using Vevo® 2100 workstation software version 3.1.0 (VisualSonics, Inc.).

Masson's trichrome staining

A total of 28 days post-surgery, all rats were euthanized with 20% CO2 for histological analysis. All heart tissues were quickly excised and dehydrated in a 30% sucrose solution. The hearts were then embedded in Tissue-Tek® O.C.T.™ Compound (Sakura Finetek USA, Inc.) and cut into 7-µm slices. Modified Masson's Trichrome Stain kit (Beijing Solarbio Science & Technology Co., Ltd.) was used to examine infarct size according to the manufacturer's instructions. A light microscope (Leica Microsystems GmbH) was used to capture images (x1 magnification). Following Masson's staining, myocardial fibers appear red, while collagen fibers appear blue. The proportion of the infarct area was quantified using Imaging Pro Plus software 6.0 (Media Cybernetics, Inc.) and calculated as follows: Infarct heart area (%)=(endocardial length + epicardial length of the infarcted area/endocardial length + epicardial length of whole left ventricle) x100%.

Statistical analysis

All data are presented as the mean ± SEM. All the cell experiments were repeated ≥ three times. GraphPad Prism 6.0 software (GraphPad Software, Inc.) was used for graph generation and data analysis. Student's t-test was performed to analyze differences between two groups. Differences among > three groups were analyzed using one-way ANOVA followed by Tukey's multiple comparisons post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

ON improves cell viability in H2O2-induced H9C2 cardiomyocyte injury

As indicated by the results of CCK-8 assays, H2O2 induced a concentration-dependent reduction in H9C2 cardiomyocyte viability compared with that in the control group (Fig. 1A). Due to its ability to induce a 50% decrease in cell viability, the 400 µmol/l dose of H2O2 was selected for subsequent experiments. Treatment with suitable concentrations of ON (5, 10 or 30 µmol/l) significantly ameliorated the H2O2-induced reductions in H9C2 cell viability (Fig. 1B) with 10 µmol/l ON exerting the optimal protective effects. Therefore, 10 µmol/l ON was chosen for subsequent experiments. An excess of ON (60 µmol/l) did not induce significant protective effects (Fig. 1B). To rule out the possibility that ON may increase the viability of the cardiomyocytes without H2O2 treatment, ON (10 µmol/l) was added to the H9C2 cell culture medium but did not affect cell viability (Fig. 1C). These results indicate the cardioprotective effects of ON against H2O2-induced H9C2 cell injury.

ON alleviates apoptosis in H2O2-induced H9C2 cardiomyocytes

Flow cytometry assays (Fig. 2A, B and D) demonstrated that H2O2 (400 µmol/l) could induce apoptosis in H9C2 cardiomyocytes. The apoptosis-promoting effect of H2O2 was confirmed by results from the western blot assays, which was evidenced by the significant increase in cleaved-caspase 3 levels (Fig. 2E and F) and significant downregulation in the Bcl2/Bax ratio (Fig. 2G and H) compared with those in the control group. After ON administration (10 µmol/l), compared with those in the H2O2 group, both the number of apoptotic cells (Fig. 2C and D) and the level of apoptosis-associated protein cleaved-caspase 3 were significantly decreased (Fig. 2E and F), whilst the Bcl2/Bax ratio was significantly increased (Fig. 2G and H). These results suggest that ON may alleviate apoptosis to promote cardiomyocyte viability.

Ononin suppresses H2O2-induced H9C2 cardiomyocyte apoptosis by enhancing autophagy

Autophagy serves an important role in cardiomyocyte injury and restoration in several cellular models (17). To determine the effects of ON on autophagy in H2O2-treated H9C2 cardiomyocytes, the expression of autophagy-associated proteins LC3-II and p62 was estimated after ON treatment. Compared with that in the H2O2 group, the H2O2 + ON group showed a significant decrease in p62 expression (Fig. 3A and B) and a significant increase in LC3-II expression (Fig. 3A and C), suggesting that ON can enhance autophagy in H2O2-treated H9C2 cells. Moreover, CQ (10 µM), an autophagy inhibitor, significantly increased the levels of p62 and LC3 II compared with those in the H2O2 + ON group (Fig. 3A-C). Furthermore, compared with that in the H2O2 + ON group and the H2O2 group, CQ significantly reversed the anti-apoptotic effects of ON (Fig. 3D), further suggesting the autophagy modulatory role of ON in the cardiomyocytes following H2O2 challenge.

ON promotes autophagy via the AMPK/mTOR pathway

The AMPK/mTOR signaling pathway serves an important role in autophagy (18). Therefore, it was hypothesized that ON may also engage autophagy through this pathway. CC, a specific AMPK inhibitor, was used to test the present hypothesis in H9C2 cells. Compared with that in the H2O2 group, ON treatment significantly increased the p-AMPK/AMPK ratio (Fig. 4A and B) whilst significantly diminishing the p-mTOR/mTOR ratio (Fig. 4A and C). After treatment with CC (10 µM) in the H2O2 group, compared with those in the H2O2 + ON group, the effects of ON on p-AMPK/AMPK (Fig. 4A and B), p-mTOR/mTOR (Fig. 4A and C), p62 (Fig. 4A and D) and LC3-II (Fig. 4A and E) were significantly reversed. However, the protective effects of ON on cell viability against H2O2 were lost after the AMPK/mTOR pathway was inhibited by CC (Fig. 4F).

Subsequently, the p-AMPK/AMPK ratio was also measured in the rat heart tissues. Compared with that in the MI group, ON significantly increased the p-AMPK/AMPK ratio, consistent with the in vitro results (Fig. 4G and H). These results suggest that ON can enhance autophagy through the p-AMPK/mTOR signaling pathway to improve viability and alleviate apoptosis.

ON treatment results in recovery of cardiac function after MI

To assess whether the anti-apoptotic effect of ON in vitro could result in favorable cardiac recovery in vivo, echocardiography was performed 28 days after MI induction. Compared with that in the MI group, ON delivery significantly improved cardiac function, as evidenced by the significantly improved LVEF and LVFS (Fig. 5A and B). To verify the improvement in cardiac function induced by ON, the heart tissue sections were examined histologically on day 28 after MI induction by Masson's trichrome staining to visualize the infarct size (Fig. 5C). Compared with that in the MI group, the ON group showed a significantly decreased infarct size (Fig. 5D). These results supported the notion that ON treatment could reduce the infarct area to preserve function.

Discussion

Stress-induced apoptosis contributes to cardiomyocyte loss (19). Therefore, inhibiting and reversing this process may prove to be an effective way to treat heart diseases (20,21). In the present study, the potential effects of ON in H2O2-induced H9C2 cardiomyocyte apoptosis were assessed. Increasing attention is being paid on exploring naturally-occurring extracts for drug development due to minimal side effects (22). As a dietary nutrient, isoflavones are widely abundant in legumes and exhibit antioxidant, anti-inflammatory and immunoregulatory effects (13). ON is a major bioactive isoflavone that can regulate oxidative stress, inflammation and immunity (15,23,24). Because of the close associations among oxidative stress, inflammation, immunity and apoptosis, it was speculated that ON can serve a role in H2O2-induced H9C2 cell injury (25). In the present study, H2O2 reliably induced H9C2 cardiomyocyte apoptosis, which was previously demonstrated (26). Following ON treatment, cell viability improved, and the apoptosis rate decreased, indicating the cardioprotective effects of ON in oxidative stress-induced cardiomyocyte apoptosis. Furthermore, ON increased the ejection fraction and decreased cardiac fibrosis in rats with MI confirmed its potential cardioprotective function.

To the best of our knowledge, the anti-apoptotic role of ON in cardiomyocytes has not been previously reported. It was speculated that autophagy may be involved in this process. Autophagy is involved in different models of cell death, including apoptosis, pyroptosis, necroptosis and necrosis (27,28). Emerging evidence has indicated that autophagy can regulate the apoptosis signaling pathway (29). Although autophagy can initiate apoptosis in cells exposed to an external stimuli, this does not mean that increased autophagy is harmful. Reducing autophagic levels can decrease apoptosis whereas and cell viability can also be improved by increasing autophagy (30). Therefore, autophagy is important for the maintenance of homeostasis and is beneficial to cells at appropriate operation levels. For example, a previous study reported that increased autophagy can promote nerve cell survival under nutrient deficiency (31). The present results demonstrated that ON increased autophagy, as observed by increased expression levels of the autophagy-associated protein markers p62 and LC3-II. Moreover, after treatment with the autophagy inhibitor CQ, ON lost its cardioprotective effects, as indicated by results from the CCK-8 assays. This suggests that ON alleviated H9C2 cardiomyocyte apoptosis at least partially by promoting autophagy.

The full signaling pathway profile that can regulate autophagy remain poorly understood, despite this process being precisely modulated (32). As a key molecule, mTOR integrates upstream signaling to inhibit the induction of autophagy (33). PI3K/AKT and MAPK/ERK1/2 signaling can activate the mTOR pathway to inhibit autophagy, whereas AMPK and p53 signaling negatively regulate mTOR to promote autophagy (34,35). The increased LC3-II and reduced p62 expression after ON treatment indicated that autophagy was upregulated, which was caused by the downregulation of mTOR phosphorylation. The present study also quantified the expression of upstream signaling components (p-AKT) of p-mTOR (data not shown). However, only p-AMPK levels were firmly enhanced after ON treatment. The benefits of p-AMPK stimulation have been previously observed in many cardiovascular diseases, including MI and ischemia reperfusion injury (36-38). In the present study, AMPK activation inhibited the phosphorylation of mTOR, leading to an increase in autophagy. After treatment with the p-AMPK inhibitor CC, ON lost its protective effects as demonstrated by the CCK-8 assays, suggesting that ON can improve cell viability and reduce apoptosis through the AMPK/mTOR/autophagy signaling pathway after H2O2-induced cardiomyocyte injury. However, the protective effect of ON on cardiomyocytes and its potential mechanisms was only reported in vitro in the present study. The functional properties of ON should be studied further in vivo before it can be applied for clinical use. By developing a MI model and evaluating the role of ON in cardiac function and myocardial fibrosis, the present study determined the protective effects of ON further to provide a novel approach for the clinical treatment of MI.

In conclusion, the present study identified a protective role of ON on cardiomyocytes and cardiac function. This effect may be mediated through the AMPK/mTOR/autophagy pathway. However, the specific mechanisms of ON in regulating cardiac function in vivo require further exploration.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the Zhejiang Medical Science and Technology Project (grant no. 2020KY290).

Availability of data and materials

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

Authors' contributions

RP designed the experiments and wrote the manuscript. QZ and JW performed the experiments and analyzed the data. RP and QZ confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The study was approved by the Animal Care and Use Committee of Wenzhou Medical University (Cixi, China; approval no. 2019635).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Okwuosa IS, Lewsey SC, Adesiyun T, Blumenthal RS and Yancy CW: Worldwide disparities in cardiovascular disease: Challenges and solutions. Int J Cardiol. 202:433–440. 2016.PubMed/NCBI View Article : Google Scholar

2 

Miao C, Lei M, Hu W, Han S and Wang Q: A brief review: The therapeutic potential of bone marrow mesenchymal stem cells in myocardial infarction. Stem Cell Res Ther. 8(242)2017.PubMed/NCBI View Article : Google Scholar

3 

Aljakna A, Fracasso T and Sabatasso S: Molecular tissue changes in early myocardial ischemia: From pathophysiology to the identification of new diagnostic markers. Int J Legal Med. 132:425–438. 2018.PubMed/NCBI View Article : Google Scholar

4 

Saha S, Panigrahi DP, Patil S and Bhutia SK: Autophagy in health and disease: A comprehensive review. Biomed Pharmacother. 104:485–495. 2018.PubMed/NCBI View Article : Google Scholar

5 

Levine B and Kroemer G: Autophagy in the pathogenesis of disease. Cell. 132:27–42. 2008.PubMed/NCBI View Article : Google Scholar

6 

Wu D, Zhang K and Hu P: The role of autophagy in acute myocardial infarction. Front Pharmacol. 10(551)2019.PubMed/NCBI View Article : Google Scholar

7 

Woodall BP and Gustafsson ÅB: Autophagy-A key pathway for cardiac health and longevity. Acta Physiol (Oxf). 223(e13074)2018.PubMed/NCBI View Article : Google Scholar

8 

Munson MJ and Ganley IG: MTOR, PIK3C3, and autophagy: Signaling the beginning from the end. Autophagy. 11:2375–2376. 2015.PubMed/NCBI View Article : Google Scholar

9 

Al-Bari MAA and Xu P: Molecular regulation of autophagy machinery by mTOR-dependent and -independent pathways. Ann NY Acad Sci. 1467:3–20. 2020.PubMed/NCBI View Article : Google Scholar

10 

Sciarretta S, Forte M, Frati G and Sadoshima J: New insights into the role of mTOR signaling in the cardiovascular system. Circ Res. 122:489–505. 2018.PubMed/NCBI View Article : Google Scholar

11 

Jia J, Bissa B, Brecht L, Allers L, Choi SW, Gu Y, Zbinden M, Burge MR, Timmins G, Hallows K, et al: AMPK, a regulator of metabolism and autophagy, is activated by lysosomal damage via a novel galectin-directed ubiquitin signal transduction system. Mol Cell. 5:951–969. 2020.PubMed/NCBI View Article : Google Scholar

12 

Messina M: A brief historical overview of the past two decades of soy and isoflavone research. J Nutr. 140:1350S–1354S. 2010.PubMed/NCBI View Article : Google Scholar

13 

Luo LY, Fan MX, Zhao HY, Li MX, Wu X and Gao WY: Pharmacokinetics and bioavailability of the isoflavones formononetin and ononin and their in vitro absorption in ussing chamber and caco-2 cell models. J Agric Food Chem. 66:2917–2924. 2018.PubMed/NCBI View Article : Google Scholar

14 

Mu H, Bai YH, Wang ST, Zhu ZM and Zhang YW: Research on antioxidant effects and estrogenic effect of formononetin from trifolium pratense (red clover). Phytomedicine. 16:314–319. 2009.PubMed/NCBI View Article : Google Scholar

15 

Dong L, Yin L, Zhang Y, Fu X and Lu J: Anti-inflammatory effects of ononin on lipopolysaccharide-stimulated RAW 264.7 cells. Mol Immunol. 83:46–51. 2017.PubMed/NCBI View Article : Google Scholar

16 

Sun Q, Kang Z, Cai J, Liu W, Liu Y, Zhang JH, Denoble PJ, Tao H and Sun X: Hydrogen-rich saline protects myocardium against ischemia/reperfusion injury in rats. Exp Biol Med (Maywood). 234:1212–1219. 2009.PubMed/NCBI View Article : Google Scholar

17 

Li DL and Hill JA: Cardiomyocyte autophagy and cancer chemotherapy. J Mol Cell Cardiol. 71:54–61. 2014.PubMed/NCBI View Article : Google Scholar

18 

Wang Y, Liu Z, Shu S, Cai J, Tang C and Dong Z: AMPK/mTOR signaling in autophagy regulation during cisplatin-induced acute kidney injury. Front Physiol. 11(619730)2020.PubMed/NCBI View Article : Google Scholar

19 

Ganguly S, Mitra A and Sarkar S: Role of α-crystallin B in regulation of stress induced cardiomyocyte apoptosis. Cardiovasc Hematol Agents Med Chem. 12:60–65. 2014.PubMed/NCBI View Article : Google Scholar

20 

Del Re DP, Amgalan D, Linkermann A, Liu Q and Kitsis RN: Fundamental mechanisms of regulated cell death and implications for heart disease. Physiol Rev. 99:1765–1817. 2019.PubMed/NCBI View Article : Google Scholar

21 

Broughton KM, Wang BJ, Firouzi F, Khalafalla F, Dimmeler S, Fernandez-Aviles F and Sussman MA: Mechanisms of cardiac repair and regeneration. Circ Res. 122:1151–1163. 2018.PubMed/NCBI View Article : Google Scholar

22 

Lundstrom K: Unlocking the therapeutic potential of plant extracts. Future Med Chem. 8:245–248. 2016.PubMed/NCBI View Article : Google Scholar

23 

Luo L, Zhou J, Zhao H, Fan M and Gao W: The anti-inflammatory effects of formononetin and ononin on lipopolysaccharide-induced zebrafish models based on lipidomics and targeted transcriptomics. Metabolomics. 15(153)2019.PubMed/NCBI View Article : Google Scholar

24 

Choy KW, Murugan D, Leong XF, Abas R, Alias A and Mustafa MR: Flavonoids as natural anti-inflammatory agents targeting nuclear factor-Kappa B (NFκB) signaling in cardiovascular diseases: A mini review. Front Pharmacol. 10(1295)2019.PubMed/NCBI View Article : Google Scholar

25 

Wu MY, Li CJ, Hou MF and Chu PY: New insights into the role of inflammation in the pathogenesis of atherosclerosis. Int J Mol Sci. 22(2034)2017.PubMed/NCBI View Article : Google Scholar

26 

Li Y, Xia J, Jiang N, Xian Y, Ju H, Wei Y and Zhang X: Corin protects H2O2-induced apoptosis through PI3K/AKT and NF-κB pathway in cardiomyocytes. Biomed Pharmacother. 97:594–599. 2018.PubMed/NCBI View Article : Google Scholar

27 

D'Arcy MS: Cell death: A review of the major forms of apoptosis, necrosis and autophagy. Cell Biol Int. 43:582–592. 2019.PubMed/NCBI View Article : Google Scholar

28 

Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, et al: Molecular mechanisms of cell death: Recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 25:486–541. 2018.PubMed/NCBI View Article : Google Scholar

29 

Ryter SW, Mizumura K and Choi AMK: The impact of autophagy on cell death modalities. Int J Cell Biol. 2014(502676)2014.PubMed/NCBI View Article : Google Scholar

30 

Wang K: Autophagy and apoptosis in liver injury. Cell Cycle. 14:1631–1642. 2015.PubMed/NCBI View Article : Google Scholar

31 

Chung KW and Chung HY: The effects of calorie restriction on autophagy: Role on aging intervention. Nutrients. 11(2923)2019.PubMed/NCBI View Article : Google Scholar

32 

Glick D, Barth S and Macleod KF: Autophagy: Cellular and molecular mechanisms. J Pathol. 221:3–12. 2010.PubMed/NCBI View Article : Google Scholar

33 

Wang Y and Zhang H: Regulation of autophagy by mTOR signaling pathway. Adv Exp Med Biol. 1206:67–83. 2019.PubMed/NCBI View Article : Google Scholar

34 

Coward J, Ambrosini G, Musi E, Truman JP, Haimovitz-Friedman A, Allegood JC, Wang E, Merrill AH Jr and Schwartz GK: Safingol (L-threo-sphinganine) induces autophagy in solid tumor cells through inhibition of PKC and the PI3-kinase pathway. Autophagy. 5:184–193. 2009.PubMed/NCBI View Article : Google Scholar

35 

Mrakovcic M and Fröhlich LF: p53-mediated molecular control of autophagy in tumor cells. Biomolecules. 8(14)2018.PubMed/NCBI View Article : Google Scholar

36 

Lee SY, Ku HC, Kuo YH, Chiu HL and Su MJ: Pyrrolidinyl caffeamide against ischemia/reperfusion injury in cardiomyocytes through AMPK/AKT pathways. J Biomed Sci. 22(18)2015.PubMed/NCBI View Article : Google Scholar

37 

Lei X, Wu Q, Leng W, Wu M, Chen L and Liang Z: Exenatide reduces cardiomyocyte apoptosis by stimulating adiponectin secretion and activating APPL1-AMPK-PPARα axis. Ann Transl Med. 7(326)2019.PubMed/NCBI View Article : Google Scholar

38 

Li R, Liu Y, Shan YG, Gao L, Wang F and Qiu CG: Bailcalin protects against diabetic cardiomyopathy through keap1/Nrf2/AMPK-mediated antioxidative and lipid-lowering effects. Oxid Med Cell Longev. 2019(3206542)2019.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

November-2021
Volume 22 Issue 5

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pan R, Zhuang Q and Wang J: Ononin alleviates H<sub>2</sub>O<sub>2</sub>‑induced cardiomyocyte apoptosis and improves cardiac function by activating the AMPK/mTOR/autophagy pathway. Exp Ther Med 22: 1307, 2021
APA
Pan, R., Zhuang, Q., & Wang, J. (2021). Ononin alleviates H<sub>2</sub>O<sub>2</sub>‑induced cardiomyocyte apoptosis and improves cardiac function by activating the AMPK/mTOR/autophagy pathway. Experimental and Therapeutic Medicine, 22, 1307. https://doi.org/10.3892/etm.2021.10742
MLA
Pan, R., Zhuang, Q., Wang, J."Ononin alleviates H<sub>2</sub>O<sub>2</sub>‑induced cardiomyocyte apoptosis and improves cardiac function by activating the AMPK/mTOR/autophagy pathway". Experimental and Therapeutic Medicine 22.5 (2021): 1307.
Chicago
Pan, R., Zhuang, Q., Wang, J."Ononin alleviates H<sub>2</sub>O<sub>2</sub>‑induced cardiomyocyte apoptosis and improves cardiac function by activating the AMPK/mTOR/autophagy pathway". Experimental and Therapeutic Medicine 22, no. 5 (2021): 1307. https://doi.org/10.3892/etm.2021.10742