Open Access

Glycyrrhizic acid exerts protective effects against hypoxia/reoxygenation‑induced human coronary artery endothelial cell damage by regulating mitochondria

  • Authors:
    • Quan Tang
    • Yuping Cao
    • Wei Xiong
    • Xixian Ke
    • Jian Zhang
    • Yu Xia
    • Daxing Liu
  • View Affiliations

  • Published online on: April 15, 2020     https://doi.org/10.3892/etm.2020.8668
  • Pages: 335-342
  • Copyright: © Tang 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

Hypoxia/reoxygenation (H/R) is one of the main causes of coronary artery disease (CAD), which is primarily induced by damage to coronary artery endothelial cells (CAECs). Glycyrrhizic acid (GA) is a natural and abundant pentacyclic triterpenoid glycoside of the licorice root extract, and it has been reported to elicit protective effects against hypoxia, inflammation and apoptosis in ischemic myocardium; therefore, GA may serve as a promising therapeutic agent for ischemia‑associated CAD. In the present study, the protective effects of GA against H/R‑induced injury in CAECs were investigated. Treatment with GA during H/R maintained cell viability and decreased H/R‑induced cell apoptosis in human CAECs. In addition, H/R‑mediated induction of intracellular and mitochondrial reactive oxygen species (ROS) was significantly decreased by GA exposure. Similar to ROS scavengers, GA treatment not only exhibited protective effects, but also maintained the mitochondrial membrane potential after H/R and inhibited H/R‑induced mitochondrial dysfunction, including deficits in ATP synthesis, mitochondrial DNA copy number and mitochondrial transcriptional activity. Furthermore, GA decreased autophagy/mitophagy, and its protective effect against H/R was abolished by autophagy promotion. Collectively, the results suggested that GA exhibited protective effects against H/R‑induced CAEC injury by decreasing ROS accumulation and maintaining mitochondrial homeostasis. Further investigation into the precise mechanisms underlying the decrease in ROS accumulation induced by GA is required.

Introduction

Ischemia/reperfusion (I/R) and hypoxia/reoxygenation (H/R) injuries are harmful to endothelial cells in the arterial system, and are the primary causes of cardiac artery disease (CAD) (1). Reperfusion and reoxygenation induce reactive oxygen species (ROS) accumulation and mitochondrial dysfunction, leading to apoptotic cell death (2). Therefore, therapeutic drugs and molecules that decrease or prevent reperfusion/reoxygenation injury have been investigated, which has improved the knowledge of the pathophysiology of the processes and has exhibited encouraging results in preclinical trials (3,4).

Glycyrrhizic acid (GA), a triterpene saponin glycoside, is the primary bioactive component of the root extract of Glycyrhiza glabra (licorice) (5). Although it is clinically used as an antiulcer, antiallergic, antioxidant, antiviral and anticancer agent (6), several studies have also demonstrated its potential protective effects against I/R- and H/R-induced endothelial injury (6,7). Cai et al (8), reported that GA elicits protective effects against myocardial I/R injury by regulating oxidative stress and inflammatory reactions via the transcriptional modification of high-mobility group box 1 and mitogen-activated protein kinase in rats (8). However, whether GA exhibits protective effects against I/R- and H/R-induced injury in coronary artery endothelial cells (CAECs) is not completely understood.

Mitochondria are essential eukaryotic organelles that are the primary source of cellular energy and participate in essential physiological processes, including cell signaling and apoptosis (5-7). During I/R or H/R, ROS accumulation decreases the mitochondrial permeability transition, decreases the mitochondrial membrane potential and alters mitochondrial homeostasis, which particularly affects myocardial and endothelial cells (9). The accumulation of excessive ROS critically damages mitochondria, resulting in damage to DNA, lipids and proteins (10). Damaged mitochondria subsequently undergo mitophagy, which results in decreased ATP production, impaired calcium buffering and ultimately, apoptosis (4,11). ROS accumulation also promotes autophagy/mitophagy to remove the damaged mitochondria, leading to further mitochondrial dysfunction (12), which has been reported to be closely associated with H/R-induced CAD.

To evaluate the effects of GA, a model of H/R injury was constructed with human CAECs (HCAECs) using a hypoxia/reoxygenation system. The present study aimed to investigate whether GA affected ROS accumulation and subsequent mitochondrial dysfunction; therefore, indicating whether GA may display protective effects against H/R-induced CAD.

Materials and methods

Cell culture and establishment of the H/R model

HCAECs were purchased from iCell Bioscience, Inc. (www.icellbioscience.com/cellDetail/914/0/-1) and cultured in Endothelial Cell Medium (Thermo Fisher Scientific, Inc.) supplemented with 10% FBS (Thermo Fisher Scientific, Inc.) at 37˚C with 5% CO2.

To mimic ischemia, hypoxia should be induced in oxygen-free and nutrition-free conditions; therefore, HCAECs were cultured with pure nitrogen for 30 min at 37˚C to expel the air and subsequently, pure nitrogen gas was used to fill the culture vessels and hypoxia chamber (Corning Inc.). Subsequently, HCAECs were cultured in hypoxic solution in the hypoxia chamber for 4 h at 37˚C. Endothelial cell medium (cat. no. CC-3162; Lonza Group, Ltd.) containing 10% FBS (cat. no. 10099; Thermo Fisher Scientific, Inc.) was pre-maintained in a hypoxia chamber at 37˚C for 24 h. Following hypoxia induction, the medium was replaced with oxygenated culture medium supplemented with 10% FBS and the culture vessels were transferred into a normoxic incubator at 37˚C with 5% CO2 for 2 h of reoxygenation. To evaluate the effect of GA on H/R, 50, 100, 150 or 200 µM GA was added to culturing medium immediately after H/R exposure and incubated for 1 h at 37˚C. After 4, 8 or 12 h, cell viability was measured by performing CCK-8 assay as described below.

To scavenge total ROS, 10 µM NAC was added into endothelial cell medium containing 10% FBS together with H/R exposure. To scavenge mitochondrial ROS, 1 µM MitoQ10 (Sigma-Aldrich; Merck KGaA) was added into medium together with H/R exposure.

To inhibit autophagy/mitophagy, 100 µmol/l rapamycin was added into endothelial cell medium containing 10% FBS together with H/R exposure. To inhibit LC3B-II degradation, 20 µmol/l chloroquine (Sigma-Aldrich; Merck KGaA) was added into endothelial cell medium containing 10% FBS together with H/R exposure.

Evaluation of cell viability

Cells (5x103) were seeded into a 96-well plate and incubated overnight 37˚C. To evaluate cell viability after hypoxia treatment for 4, 8 or 12 h, 10 µl Cell Counting Kit-8 (CCK-8) solution (Shanghai Shenggong Biology Engineering Technology Serve, Ltd.) was added to each well and incubated at 37˚C for 2 h. The absorbance of each well was measured at a wavelength of 450 nm using a Multiskan spectrum microplate reader (Thermo Fisher Scientific, Inc.).

Apoptosis assay

Apoptotic cells were detected using the Annexin V/PI Apoptosis kit (BioVision, Inc.) following the manufacturer's protocol. Briefly, cell suspensions were made using 0.25% trypsin and the cell concentration was modified to 1x106 cells/ml. Subsequently, cells (5x105) were stained with 5 µl Annexin V-FITC and 10 µl propidium iodide (PI) for 10 min in darkness at room temperature. Apoptotic cells were detected using a 3 laser Navios flow cytometer (Beckman Coulter, Inc.). The Annexin V-FITC positive/PI negative (early stage apoptosis) and Annexin V-FITC positive/PI positive (late stage apoptosis) subpopulations were considered as apoptotic cells. Data were analyzed using FlowJo software (version 10.5.2; FlowJo LLC).

Detecting intracellular and mitochondrial ROS levels in HCAECs

To measure intracellular and mitochondrial ROS levels, 2',7'-dichlorofluorescin diacetate (DCFH-DA) and MitoSOX™ staining were performed, respectively. Nuclei were counterstained with DAPI to a final concentration of 5 µg/ml (Beyotime Institute of Biotechnology) at room temperature for 5 min. Cells were washed 3 times with ice-cold PBS. Subsequently, cells were incubated with 1 ml serum-free medium containing 10 µM DCFH-DA probe or 5 µM MitoSOX™ in the dark at 37˚C for 30 min with gentle shaking every 5 min. Cells were washed 3 times with ice-cold PBS. Green fluorescence was observed using an X71 (U-RFL-T) fluorescence microscope (Olympus Corporation) at an excitation wavelength of 488 nm and magnification, x40.

JC-1 staining

JC-1 staining (Thermo Fisher Scientific, Inc.) was used to investigate the mitochondrial membrane potential. Cells were stained with 15 µg/ml JC-1 at 37˚C for 30 min in dark. Subsequently, cells were washed 3 times with PBS. Stained cells were observed using an X71 (U-RFL-T) fluorescence microscope (Olympus Corporation) at a magnification, x100.

ATP measurement

To assess ATP production, the ATP Bioluminescent assay kit (Sigma-Aldrich; Merck KGaA) was used according to the manufacturer's protocol. Cells were washed 3 times with ice-cold PBS and subsequently lysed using a reaction buffer containing 0.22 M sucrose, 0.12 M mannitol, 40 mM tricine (pH 7.5) and 1 mM EDTA. ATP production was measured using an Optocomp I BG-1 luminometer (GEM Biomedical, Inc.).

Measurement of telomere length and mitochondrial DNA (mtDNA) copy number

Total DNA (including nuclear and mitochondrial genomic DNA) was extracted using a TIANamp total DNA kit (Tiangen Biotech Co., Ltd.) and directly employed as a template for PCR. The mtDNA copy number was then assessed by qPCR using SYBRGreen master mix (Thermo Fisher Scientific, Inc.) and the ABI7500 Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The following primer pairs were used for qPCR: 200 nM mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 1 (ND1) forward, 5'-CCCTAAAACCCGCCACATCT-3' and reverse, 5'-GAGCGATGGTGAGAGCTAAGGT-3'; and 167 nM cytoglobin (HGB) forward, 5'-GTGCACCTGACTCCTGAGGAGA-3' and reverse, 5'-CCTTGATACCAACCTG-CCCAG-3'. The following thermocycling conditions were used for qPCR: Initial denaturation, 98˚C for 30 sec, followed by 40 cycles of 98˚C for 10 sec and 60˚C for 60 sec. mtDNA copy numbers were normalized to the nuclear single-copy gene HGB. The relative mtDNA copy number was calculated using the 2-ΔΔCq method (13).

Mitochondrial transcriptional activity

Total RNA was extracted from cells using TRIzol® reagent (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Total RNA was reverse transcribed into cDNA using the RT-for-PCR kit (Qiagen, Inc.) in accordance with the manufacturer's protocol. Subsequently, qPCR was performed using SYBRGreen master mix (Thermo Fisher Scientific, Inc.) and the ABI7500 Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The following primer pairs were used for qPCR: Mitochondrially encoded cytochrome c oxidase I forward, 5'-ATGCGGCCATAGGTTCTGC-3' and reverse, 5'-TCCTCAAGATGTCTCAGTTCCAT-3'; ND1 forward, 5'-TCGTCATAATCTGTCCCTACACA-3' and reverse, 5'-CGGCTTCGGCTCTTAGCAAA-3'; β-actin forward, 5'-CATGTACGTTGCTATCCAGGC3' and reverse, 5'-CTCCTTAATGTCACGCACGAT-3'. The following thermocycling conditions were used for qPCR: Initial denaturation at 98˚C for 30 sec, followed by 45 cycles of 98˚C for 10 sec and 60˚C for 60 sec. mRNA expression levels were normalized to the internal reference gene β-actin. Relative expression levels were calculated using 2-ΔΔCq method (13).

Western blotting

Total protein was extracted using the SoniConvert™ system (DocSense) and RIPA buffer (Sigma-Aldrich; Merck KGaA). Total protein was denatured at 100˚C for 10 min and was quantitatively measured using a bicinchoninic acid assay kit (Sigma-Aldrich; Merck KGaA) in accordance with the manufacturer's protocol. Total protein (20 µg) were separated by 8-16% SDS-PAGE and transferred to nitrocellulose membranes. After blocking in 5% bovine serum albumin (Sigma-Aldrich; Merck KGaA) at room temperature for 30 min, the membranes were incubated with primary antibodies targeted against microtubule associated protein 1 light chain 3B (LC3B; cat. no. ab48394; 1:1,000; Abcam), p62 (cat. no. ab109012; 1:2,000; Abcam) and β-actin (cat. no. ab8226; 1:5,000; Abcam) at room temperature for 1 h. After three washes with PBS-T, membranes were incubated with horseradish peroxidase-conjugated goat anti-rabbit secondary antibodies (cat. no. ab7090; 1:5,000; Abcam) at room temperature for 1 h. Protein bands were visualized using a chemiluminescence kit (Thermo Fisher Scientific, Inc.). Protein expression levels were quantified using Image J software (version 1.51; National Institutes of Health) with β-actin as the loading control.

Statistical analysis

Data are presented as the mean ± standard error of the mean and analyzed using SPSS software (version 16.0; SPSS, Inc.) All experiments were repeated 3 times independently. Comparisons among multiple groups were analyzed using one-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

GA exerts protective effects against H/R-induced cellular injury in HCAECs

To evaluate the effect of GA on H/R-induced cellular injury in HCAECs, different concentrations of GA (0-200 µM) were added to the medium during H/R induction, and the CCK-8 assay was subsequently performed after 4, 8 or 12 h. The results suggested that 100, 150 and 200 µM GA significantly decreased H/R-induced cell injury at each time point compared with the 0 µM GA group (Fig. 1A). At each time point, there were no significant differences between the protective effects of 100 and 200 µM GA; therefore, 100 µM GA was used for subsequent experiments.

Considering that H/R induces cell injury and decreases cell viability mainly by inducing apoptosis (13), apoptotic cell death following GA incubation was assessed at different time points. GA incubation significantly decreased the Annexin V-FITC+/PI- and Annexin V-FITC+/PI+ subpopulations at each time point compared with the H/R group (Fig. 1B). The highest proportion of apoptotic cells was observed at 4 h after H/R induction, indicating that H/R-induced cell injury peaked at this time point.

GA decreases total and mitochondrial ROS levels and consequently decreases H/R-induced apoptosis

Considering that H/R induces intracellular and mitochondrial ROS accumulation (14-17), whether GA altered H/R-induced ROS accumulation was investigated and the rate of apoptosis was assessed. By detecting intracellular ROS (with DCFH-DA labeling) and mitochondrial ROS (with MitoSOX™ Red), total ROS and mitochondrial ROS levels after GA treatment were measured. H/R-induced ROS accumulation was significantly decreased by incubation with 100 µM GA (Fig. 2A). In addition, H/R-induced mitochondrial ROS accumulation was decreased by incubation with 100 µM GA, which exhibited similar effects compared with treatment with 1 µM MitoQ10 (Fig. 2B), indicating that GA and MitoQ10 elicited similar effects.

Subsequently, the effects of GA on cell viability and apoptosis were measured. A CCK-8 assay was performed and the results revealed that GA treatment significantly increased the H/R-induced reduction of cell viability, which was an effect similar to that of NAC and MitoQ10 treatments (Fig. 2C). Annexin-FITC/PI double staining followed by flow cytometry was performed to detect apoptosis. Treatment with GA, N-acetylcysteine (NAC) or MitoQ10 decreased H/R-induced cell apoptosis (Fig. 2D). The results indicated that H/R-induced ROS accumulation was important for inducing apoptosis, and that GA treatment potentially displayed a protective effect against cell apoptosis by decreasing ROS accumulation.

GA treatment maintains the mitochondrial membrane potential and mitochondrial function

To investigate the effects of H/R-induced ROS accumulation on the mitochondrial membrane potential, JC-1 staining was performed. The mitochondrial membrane potential was notably decreased following H/R induction, as suggested by a decrease in red fluorescence compared with the normoxia group (Fig. 3A). Treatment with 100 µM GA, 10 µM NAC or 1 µM MitoQ10 markedly increased red fluorescence compared with the H/R group, which indicated that the 3 treatments maintained the mitochondrial membrane potential following H/R induction. To further investigate mitochondrial function, ATP synthesis, mtDNA copy number and mitochondrial transcriptional activity were assessed (Fig. 3B-D). Treatment with GA or NAC significantly protected cells against H/R-induced mitochondrial dysfunction, decreasing ATP synthesis and normalizing transcriptional activity to that of the internal reference gene (β-actin). In addition, the relatively high mtDNA copy number following GA treatment was normalized to HGB, which is a nuclear single-copy gene. The results revealed that GA treatment was potentially associated with promotion of mitochondria biogenesis or inhibition of mitophagy.

GA decreases autophagy/mitophagy and autophagy induction abolishes its protective effects

To determine whether GA treatment maintained mitochondrial function via autophagy/mitophagy, markers of autophagy/mitophagy, including LC3B and p62, were detected by western blotting. H/R-induced LC3B-II expression was significantly decreased by GA, NAC or MitoQ10 treatment. Similarly, the expression of p62, which is downregulated during autophagy/mitophagy, was also markedly maintained by treatment with GA, NAC or MitoQ10 following H/R induction (Fig. 4A).

To further clarify whether GA exerted protective effects by inhibiting autophagy/mitophagy, cell viability and apoptosis were analyzed in cells treated with GA and rapamycin, an autophagy-inducing compound. Rapamycin exposure abolished GA-induced protective effects on cell viability and apoptosis, which indicated that the protective effects of GA were associated with a decrease in autophagy/mitophagy (Fig. 4B and C).

Collectively, the results suggested that GA decreased H/R-induced ROS accumulation, maintained the mitochondrial membrane potential and inhibited mitophagy/autophagy, which protected cells against H/R-induced injury.

Discussion

In the present study, the protective effects of GA against H/R-induced decreases in cell viability and increases in cell apoptosis in HCAECs were investigated. Treatment with GA significantly inhibited H/R-induced ROS accumulation, maintained the mitochondrial membrane potential and mitochondrial homeostasis, and prevented mitochondrial dysfunction following H/R.

I/R and H/R are common causes of tissue and cell damage due to a lack of blood supply or oxygen. Unfortunately, restoration of the blood or oxygen supply leads to further oxidative damage and inflammatory effects by inducing oxidative stress (1). Certain endothelial cells in different organs are particularly sensitive to ischemia and hypoxia due to their direct contact with the circulation, and damage to these cells can result from an oxidative stress imbalance arising from the inflammatory response (18). I/R- and H/R-induced ROS accumulation damages mitochondria, decreases the mitochondrial membrane potential, damages DNA, lipids and proteins, and promotes mitophagy (16). Considering the critical effects of ROS in promoting cell injury following H/R, scavenging ROS is a promising treatment strategy. N-n-butyl haloperidol iodide (F2) has been reported to exert protective effects against myocardial I/R injury by downregulating H/R-induced ROS levels, inactivating JNK and downregulating early growth response 1 protein expression in H9c2 cells (19). Metformin, a first-line drug for the management of type 2 diabetes, has been reported to protect cardiomyocytes against H/R injury by inhibiting ROS generation and inflammatory responses (20). Collectively, the aforementioned studies indicated that GA might protect against H/R-induced injury by regulating ROS levels.

In the present study, H/R induced intracellular and mitochondrial ROS accumulation, and treatment with GA significantly decreased ROS levels. Further investigation is required to determine whether ROS generation or accumulation is affected by GA. To distinguish mitochondrial ROS from intracellular ROS, 0.5 µM MitoSOX with an excitation wavelength of 365 nm was used. H/R induced intracellular and mitochondrial ROS accumulation; however, whether the observed increase in mitochondrial ROS levels was amplified by intracellular ROS accumulation, as has been previously reported, was not investigated in the present study (21,22). Although H/R-induced intracellular ROS accumulation directly interacts with mitochondrial proteins and lipids to accelerate mitochondrial dysfunction (23), in the present study, scavengers of intracellular and mitochondrial ROS decreased the rate of apoptosis following H/R, suggesting that mitochondrial ROS directly induced apoptosis. A decrease in the percentage of Annexin V+/PI- and Annexin V+/PI+ cells following treatment with GA in H/R-induced cells was also observed, which suggested that necrosis had also occurred; however, the effects of GA on necrosis require further investigation.

An excess of mitochondrial ROS leads to mitophagy and subsequent apoptosis, which is accompanied by a decrease in ATP production and collapse of the mitochondrial membrane potential (24,25). It has also been reported that GA exerts a protective role against hypoxia-induced mitochondrial damage by regulating mitochondria (26). Following H/R, GA treatment inhibited the decrease in ATP production, the collapse of the mitochondrial membrane potential and the decrease in mtDNA copy number. By detecting the hallmarks of autophagy/mitophagy, it was further revealed that GA treatment inhibited ROS-induced autophagy/mitophagy and subsequent cell apoptosis by decreasing ROS levels. The effects of GA on the viability of cells following H/R induction was analyzed using the CCK-8 assay. Future studies should perform time-lapse live-cell experiments to reveal the effects of GA on myocardial cells and H/R-induced necrosis.

The present study had several limitations. Firstly, H/R-induced mitochondrial ROS accumulation may induce endothelial injury; however, whether the observed increase in mitochondrial ROS levels was amplified by intracellular ROS was not investigated in the present study. Secondly, the exact mechanism underlying GA-induced reductions in ROS accumulation was not identified in the present study and requires further investigation.

Acknowledgements

The authors would like to thank Mrs Yun Bai (Third Military Medical University, Chongqing) for language editing the manuscript.

Funding

The present study was supported by the Starting Scientific Foundation of Zunyi Medical University (grant no. SS20180601ZF).

Availability of data and materials

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

Authors' contributions

QT designed the study. YC, WX and XK performed the cellular experiments and analyzed the data. JZ and YX collected the data, wrote the manuscript and performed the flow cytometry experiments. DL contributed to designing the study and writing the manuscript, and also provided supervision. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Turer AT and Hill JA: Pathogenesis of myocardial ischemia-reperfusion injury and rationale for therapy. Am J Cardiol. 106:360–368. 2010.PubMed/NCBI View Article : Google Scholar

2 

Fernandez-Jimenez R, Garcia-Prieto J, Sanchez-Gonzalez J, Agüero J, López-Martín GJ, Galán-Arriola C, Molina-Iracheta A, Doohan R, Fuster V and Ibáñez B: Pathophysiology underlying the bimodal edema phenomenon after myocardial ischemia/reperfusion. J Am Coll Cardiol. 66:816–828. 2015.PubMed/NCBI View Article : Google Scholar

3 

Rezende PC, Ribas FF, Serrano CJ and Hueb W: Clinical significance of chronic myocardial ischemia in coronary artery disease patients. J Thorac Dis. 11:1005–1015. 2019.PubMed/NCBI View Article : Google Scholar

4 

Bosetti F, Brizzi F, Barogi S, Mancuso M, Siciliano G, Tendi EA, Murri L, Rapoport SI and Solaini G: Cytochrome c oxidase and mitochondrial F1F0-ATPase (ATP synthase) activities in platelets and brain from patients with Alzheimer's disease. Neurobiol Aging. 23:371–376. 2002.PubMed/NCBI View Article : Google Scholar

5 

Blackstone NW: The impact of mitochondrial endosymbiosis on the evolution of calcium signaling. Cell Calcium. 57:133–139. 2015.PubMed/NCBI View Article : Google Scholar

6 

Saraste M: Oxidative phosphorylation at the fin de siecle. Science. 283:1488–1493. 1999.PubMed/NCBI View Article : Google Scholar

7 

Wang C and Youle RJ: The role of mitochondria in apoptosis*. Annu Rev Genet. 43:95–118. 2009.PubMed/NCBI View Article : Google Scholar

8 

Cai X, Wang X, Li J and Chen S: Protective effect of glycyrrhizin on myocardial ischemia/reperfusion injury-induced oxidative stress, inducible nitric oxide synthase and inflammatory reactions through high-mobility group box 1 and mitogen-activated protein kinase expression. Exp Ther Med. 14:1219–1226. 2017.PubMed/NCBI View Article : Google Scholar

9 

Valls-Lacalle L, Barba I, Miro-Casas E, Alburquerque-Béjar JJ, Ruiz-Meana M, Fuertes-Agudo M, Rodríguez-Sinovas A and García-Dorado D: Succinate dehydrogenase inhibition with malonate during reperfusion reduces infarct size by preventing mitochondrial permeability transition. Cardiovasc Res. 109:374–384. 2016.PubMed/NCBI View Article : Google Scholar

10 

Xu M, Bi X, He X, Yu X, Zhao M and Zang W: Inhibition of the mitochondrial unfolded protein response by acetylcholine alleviated Hypoxia/Reoxygenation-Induced apoptosis of endothelial cells. Cell Cycle. 15:1331–1343. 2016.PubMed/NCBI View Article : Google Scholar

11 

Guo X, Wu J, Du J, Ran J and Xu J: Platelets of type 2 diabetic patients are characterized by high ATP content and low mitochondrial membrane potential. Platelets. 20:588–593. 2009.PubMed/NCBI View Article : Google Scholar

12 

Saito T and Sadoshima J: Molecular mechanisms of mitochondrial autophagy/mitophagy in the heart. Circ Res. 116:1477–1490. 2015.PubMed/NCBI View Article : Google Scholar

13 

Livak KD and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

14 

Cao X, Wang X, Ling Y, Song X, Yang P, Liu Y, Liu L, Wang L, Guo J and Chen A: Comparison of the degree of autophagy in neonatal rat cardiomyocytes and H9c2 cells exposed to hypoxia/reoxygenation. Clin Lab. 60:809–814. 2014.PubMed/NCBI View Article : Google Scholar

15 

Zhang Y, Shi G, Zheng J, Tang Z, Gao P, Lv Y, Guo F and Jia Q: The protective effects of N-n-butyl haloperidol iodide on myocardial ischemia-reperfusion injury in rats by inhibiting Egr-1 overexpression. Cell Physiol Biochem. 20:639–648. 2007.PubMed/NCBI View Article : Google Scholar

16 

Zhang Y, Chen G, Zhong S, Zheng F, Gao F, Chen Y, Huang Z, Cai W, Li W, Liu X, et al: N-n-butyl haloperidol iodide ameliorates cardiomyocytes hypoxia/reoxygenation injury by extracellular calcium-dependent and -independent mechanisms. Oxid Med Cell Longev. 2013(912310)2013.PubMed/NCBI View Article : Google Scholar

17 

Paradies G, Paradies V, Ruggiero FM and Petrosillo G: Mitochondrial bioenergetics and cardiolipin alterations in myocardial ischemia-reperfusion injury: Implications for pharmacological cardioprotection. Am J Physiol Heart Circ Physiol. 315:H1341–H1352. 2018.PubMed/NCBI View Article : Google Scholar

18 

Cadenas S: ROS and redox signaling in myocardial ischemia-reperfusion injury and cardioprotection. Free Radic Biol Med. 117:76–89. 2018.PubMed/NCBI View Article : Google Scholar

19 

Carden DL and Granger DN: Pathophysiology of ischaemia-reperfusion injury. J Pathol. 190:255–266. 2000.PubMed/NCBI View Article : Google Scholar

20 

Zhang Y, Liao H, Zhong S, Gao F, Chen Y, Huang Z, Lu S, Sun T, Wang B, Li W, et al: Effect of N-n-butyl haloperidol iodide on ROS/JNK/Egr-1 signaling in H9c2 cells after hypoxia/reoxygenation. Sci Rep. 5(11809)2015.PubMed/NCBI View Article : Google Scholar

21 

Hu M, Ye P, Liao H, Chen M and Yang F: Metformin protects h9c2 cardiomyocytes from high-glucose and hypoxia/reoxygenation injury via inhibition of reactive oxygen species generation and inflammatory responses: Role of AMPK and JNK. J Diabetes Res. 2016(2961954)2016.PubMed/NCBI View Article : Google Scholar

22 

Daiber A, Di Lisa F, Oelze M, Kröller-Schön S, Steven S, Schulz E and Münzel T: Crosstalk of mitochondria with NADPH oxidase via reactive oxygen and nitrogen species signalling and its role for vascular function. Br J Pharmacol. 174:1670–1689. 2017.PubMed/NCBI View Article : Google Scholar

23 

Tajeddine N: How do reactive oxygen species and calcium trigger mitochondrial membrane permeabilisation? Biochim Biophys Acta. 1860:1079–1088. 2016.PubMed/NCBI View Article : Google Scholar

24 

Liu P, Lin Y, Tang X, Zhang P, Liu B, Liu Y and Miao F: Helix B surface peptide protects cardiomyocytes against hypoxia/reoxygenation-induced apoptosis through mitochondrial pathways. J Cardiovasc Pharmacol. 67:418–426. 2016.PubMed/NCBI View Article : Google Scholar

25 

Ahn HJ, Kim KI, Kim G, Moon E, Yang SS and Lee JS: Atmospheric-pressure plasma jet induces apoptosis involving mitochondria via generation of free radicals. PLoS One. 6(e28154)2011.PubMed/NCBI View Article : Google Scholar

26 

Du JK, Cong BH, Yu Q, Wang H, Wang L, Wang CN, Tang XL, Lu JQ, Zhu XY and Ni X: Upregulation of microRNA-22 contributes to myocardial ischemia-reperfusion injury by interfering with the mitochondrial function. Free Radic Biol Med. 96:406–417. 2016.PubMed/NCBI View Article : Google Scholar

27 

Xu CL, Liang CH, Sun WX, Chen J and Chen X: Glycyrrhizic acid ameliorates myocardial ischemic injury by the regulation of inflammation and oxidative state. Drug Des Devel Ther. 12:1311–1319. 2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

July-2020
Volume 20 Issue 1

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
Tang Q, Cao Y, Xiong W, Ke X, Zhang J, Xia Y and Liu D: Glycyrrhizic acid exerts protective effects against hypoxia/reoxygenation‑induced human coronary artery endothelial cell damage by regulating mitochondria. Exp Ther Med 20: 335-342, 2020
APA
Tang, Q., Cao, Y., Xiong, W., Ke, X., Zhang, J., Xia, Y., & Liu, D. (2020). Glycyrrhizic acid exerts protective effects against hypoxia/reoxygenation‑induced human coronary artery endothelial cell damage by regulating mitochondria. Experimental and Therapeutic Medicine, 20, 335-342. https://doi.org/10.3892/etm.2020.8668
MLA
Tang, Q., Cao, Y., Xiong, W., Ke, X., Zhang, J., Xia, Y., Liu, D."Glycyrrhizic acid exerts protective effects against hypoxia/reoxygenation‑induced human coronary artery endothelial cell damage by regulating mitochondria". Experimental and Therapeutic Medicine 20.1 (2020): 335-342.
Chicago
Tang, Q., Cao, Y., Xiong, W., Ke, X., Zhang, J., Xia, Y., Liu, D."Glycyrrhizic acid exerts protective effects against hypoxia/reoxygenation‑induced human coronary artery endothelial cell damage by regulating mitochondria". Experimental and Therapeutic Medicine 20, no. 1 (2020): 335-342. https://doi.org/10.3892/etm.2020.8668