2,506
Views
1
CrossRef citations to date
0
Altmetric
Research Article

Ginsenoside Rh2 mitigates myocardial damage in acute myocardial infarction by regulating pyroptosis of cardiomyocytes

, , , &
Article: 2229536 | Received 17 Mar 2023, Accepted 20 Jun 2023, Published online: 03 Jul 2023

ABSTRACT

Background

Acute myocardial infarction (AMI) is one of the most important causes of mortality among patients with cardiovascular disease. Ginsenoside Rh2 plays a protective role in cardiovascular diseases. Furthermore, pyroptosis reportedly participates in regulating the occurrence and development of AMI. However, whether ginsenoside Rh2 contributes to mitigating AMI by regulating cardiomyocyte pyroptosis remains unknown.

Methods

In the present study, we established an AMI model in rats. Next, we determined the effects of ginsenoside Rh2 on AMI by examining the myocardial infarct area, while regulation of myocardial pyroptosis was determined by assessing related factors. We established a cardiomyocyte model using hypoxia/reoxygenation (H/R) treatment. The expression of pyroptosis-related factors was determined following ginsenoside Rh2 treatment. In addition, we evaluated the correlation between ginsenoside Rh2 and the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway at the mechanistic level.

Results

Herein, we observed that ginsenoside Rh2 alleviated AMI in rats and cells. Notably, the expression levels of inflammatory factors were reduced in AMI rats and cells. Furthermore, AMI rats and cells exhibited high expression levels of cleaved caspase-1 and gasdermin D, which were downregulated following treatment with ginsenoside Rh2. Further analysis revealed that ginsenoside Rh2 could inhibit cardiomyocyte pyroptosis by regulating the PI3K/AKT signaling pathway.

Conclusions

Collectively, the findings of the present study demonstrated that ginsenoside Rh2 regulates pyroptosis in cardiomyocytes to alleviate AMI in vivo and in vitro, thereby affording a novel therapeutic approach to treat AMI.

Background

Acute myocardial infarction (AMI) is characterized by a short period of rapid coronary occlusion that results in myocardial ischemia and necrosis in the corresponding perfusion area. Prolonged ischemia can lead to sudden cardiac death or develop into heart failure, impairing left ventricular pump function (Citation1,Citation2). The mortality rate of AMI has revealed an exponentially increasing trend with increasing patient age (Citation3). An intense inflammatory response is known to occur during myocardial infarction (MI), and this inflammatory process, while critical for tissue healing, can lead to excessive injury and dysregulation of ventricular remodeling, ultimately resulting in heart failure and impaired myocardial function (Citation4,Citation5).

Pyroptosis is a type of programmed cell death involving the inflammatory necrosis of cells (Citation6). Unlike apoptosis, pyroptosis does not depend on caspase-3 but on the continuous activation of caspase-1, with interleukin (IL)-1β, high-mobility group box 1, IL-18, and other inflammatory factors released in large quantities, resulting in inflammatory response, with cells ultimately manifesting osmotic disintegration (Citation7). Pyroptosis reportedly mediates the occurrence and development of several diseases, including infectious diseases, nervous system diseases, cardiovascular diseases (CVDs), metabolic diseases, and inflammatory immune diseases (Citation8). Pyroptosis is involved in regulating AMI development, and its molecular mechanism has been previously explored (Citation9,Citation10).

In traditional Chinese medicine, ginseng is the first choice of drug to correct the disrupted balance and is rich in several pharmacologically active ingredients, with ginsenoside deemed the most important (Citation11). Ginsenoside Rh2 can inhibit the nuclear factor (NF)-κB signaling pathway and reduce the inflammatory response by enhancing the expression of transforming growth factor (TGF)-β1 (Citation12). Ginsenoside Rh2 has been shown to afford a protective effect against CVDs. It can improve cardiac function and fibrosis by enhancing the peroxisome proliferator-activated receptor delta (PPARδ) signaling pathway and is a promising alternative therapy for cardiac fibrosis (Citation13). Furthermore, ginsenoside Rh2 can improve the inflammatory microenvironment by enhancing exosome protection against myocardial injury (Citation14). Recently, ginsenoside Rh2 was found to play an anti-inflammatory role in alleviating AMI by regulating cardiomyocyte pyroptosis in rats (Citation15). However, further in vitro and in vivo studies are required to establish its protective effects and elucidate the underlying molecular mechanisms.

In the present study, we established a rat model to examine the protective effect of ginsenoside Rh2 in AMI rats and determine its potential to regulate pyroptosis in cardiomyocytes. In addition, the protective effect of ginsenoside Rh2-induced pyroptosis was assessed in a cellular model of hypoxia/reoxygenation (H/R)-induced H9C2 cardiomyocytes. Furthermore, we elucidated the molecular mechanism by which ginsenoside Rh2 inhibits cardiomyocyte pyroptosis by regulating the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway, thereby alleviating AMI.

Methods

Grouping and modeling

To measure the possible effect of Ginsenoside Rh2 on AMI, ten-week-old male Sprague Dawley rats (approximately 6–8 weeks, 220–250 g) were randomly divided into three groups: sham, AMI and AMI+Ginsenoside Rh2. All rats were housed at 23 ± 2°C under a 12 h light/12 h dark cycle with free access to food and water. The AMI and AMI+Ginsenoside Rh2 were established as AMI model by referring to previously described (Citation16). 2,2,2-Tribromoethanol (200 mg/kg; Sigma, St. Louis, MO, USA) was intraperitoneally injected to anesthetize animals. A left thoracotomy was performed with a small incision placed at the third and fourth intercostal spaces, and a 7–0 prolene suture (Ethicon, Inc., Somerville, NJ, USA) was employed to ligate the left anterior descending artery (LAD). LAD ligation was performed for 30 min, followed by reperfusion for 4 h and intravenous injection of 2% Evans Blue via the jugular vein. Rats in the sham group underwent the same procedure without ligation. All operations were performed under aseptic conditions. The rats of AMI+Ginsenoside Rh2 group were intragastrically administered 4 mg/kg ginsenoside Rh2. As previously described, ginsenoside Rh2 powder was dissolved in dimethyl sulfoxide at a concentration of 50 mM to prepare a stock solution, and 2 µM was selected as the treatment concentration (Citation17). The experimental and animal care procedures were approved by the Affiliated Hospital of Zunyi Medical University (Approval Number. ZMU21-2207-032) and conformed to the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health.

Cell culture and H/R treatment

Rat heart-derived H9c2 cells were acquired from the cell bank of the Chinese Academy of Sciences (Shanghai, China) and cultured in Dulbecco’s Modified Eagle Medium (Hyclone, Logan, UT, USA), supplemented with 100 IU/mL penicillin – streptomycin (Beyotime, Shanghai, China) and 10% fetal bovine serum (Hyclone, Logan, UT, USA) at 37°C in a 5% CO2 incubator. H/R group: H9c2 cells were subjected to 16 h of hypoxia (O2:N2:CO2, 1:94:5), followed by 2 h of reoxygenation.

Detection of myocardial infarct area (IA)

Evans Blue and TTC staining were used to evaluate the IA of ischemia/reperfusion injury, according to previous reports (Citation18). The rat heart was washed with phosphate-buffered saline and cut into four 2 mm-thick slices from the apex to the base. After incubation with 2% TTC for 20 min, sections were divided into three areas: non-ischemic (blue), risk (area at risk [AAR]; dark red), and infarct (IA, pale white) areas. Sections of the cardiac papillary muscle plane were imaged under a microscope and quantified using Image-Pro Plus 6.0 (Media Cybernetics). The percentage of IA was calculated using the following formula: IA/total left ventricle (LV) 100%.

Determination of levels of necrotic cardiomyocytes

Following various treatments, creatine kinase (CK), malondialdehyde (MDA) and lactate dehydrogenase (LDH) levels were assessed using commercially available biochemical kits (Beijing KEMEI DONGYA Biotechnology Ltd.) The procedures were performed according to the manufacturer’s instructions.

Enzyme-linked immunosorbent assay (ELISA)

Serum concentrations of IL-18 and IL-1β in AMI rats were detected using commercial ELISA kits (Nanjing Jiancheng Bioengineering Institute, Nanjing, China), according to the manufacturer’s instructions (Citation19). The absorbance was measured at 450 nm using the color intensity development.

Quantitative reverse transcription polymerase chain reaction (Qrt-PCR)

Total RNA was extracted from cardiac tissues or treated cells using TRIzol reagent. And reversely transcribed into complementary DNA (cDNA) by a PrimeScript 1st Strand cDNA synthesis kit (Takara, Dalian, China). qRT-PCR was accomplished by a SYBR® Premix Ex Taq™ Kit (Takara) on ABI7300 real-time PCR system (Applied Biosystems, Foster City, CA, USA). The relative expression of genes was calculated by 2−ΔΔCt, and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as endogenous control. The primer sequences are detailed in .

Table 1. Primer information in this study.

Western blotting assay

Proteins were extracted from rat heart tissue and neonatal cardiomyocytes using a radioimmunoprecipitation assay solution containing protease inhibitors (Solarbio, Beijing, China) and analyzed by western blotting. Protein concentration was measured using a BCA kit (Beyotime, Shanghai, China). Proteins were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis, transferred to nitrocellulose membranes, and blocked with 5% bovine serum albumin at room temperature for 2 h. Thereafter, the membranes were incubated with antibodies against p-PI3K (cat. no. 302958), t-PI3K (cat. no. 120924), p-AKT (cat. no. 38449), t-AKT (cat. no. 126433), nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) (cat. no. 15101), cleaved caspase-1 (cat. no. 67314), pro-caspase-1 (cat. no. 93709), apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) (cat. no. 67824), and GAPDH (cat. no. TA-08) (1:1000; all antibodies were purchased from Abcam). Membranes were incubated with horseradish peroxidase-conjugated secondary antibodies at room temperature for 1 h. Protein bands were visualized using an enhanced chemiluminescence system (Thermo Fisher Scientific, Inc.).

Histological examinations

The left ventricle samples of rats in each group were soaked in 4% paraformaldehyde solution for 24 h. After fixation, the specimen was embedded in paraffin wax and cut into 5 μm thick slices on the slide. After that, H&E staining was performed and the cardiac morphology was observed using light microscopy.

Statistical analysis

Data values are expressed as mean ± standard deviation. The Student’s t-test was used for between-group comparisons. Differences among groups were analyzed using one-way analysis of variance. Differences were considered statistically significant at P < .05. All statistical analyses were performed using GraphPad Prism 8.0 (GraphPad Software, La Jolla, CA, USA).

Results

Ginsenoside Rh2 alleviated pyroptosis in AMI rats

We assessed whether ginsenoside Rh2 could exert a cardioprotective effect against AMI. The AMI group exhibited a higher percentage of MI area than the sham group, which was partially reduced following treatment with ginsenoside Rh2 (). In addition, H&E staining showed that myocardial damage scores in AMI group was more higher than that in sham group, and the damage scores was partially relieved after Ginsenoside Rh2 treatment (). Levels of MDA, LDH, CK, IL-1β and IL-18 were increased in AMI rats, which were effectively decreased following treatment with ginsenoside Rh2 (). In addition, mRNA levels of IL-1β, IL-18, caspase-1, NLRP3, ASC and gasdermin D (GSDND) were increased in AMI rats (); And protein levels of cleaved caspase-1, NLRP3, ASC, and GSDND were increased in AMI rats. However, these mRNA and protein levels were effectively decreased after treatment with ginsenoside Rh2 ().

Figure 1. Treatment with ginsenoside Rh2 alleviates AMI in rats.

(a) Treatment with ginsenoside Rh2 partially reduces the myocardial infarction area. (b) HE staining assesses histology of heart tissue. Scale bar indicates 50μm. (c-e) Treatment with ginsenoside Rh2 partially reduces changes in expression levels of MDA, LDH and CK in AMI rats. (f) Treatment with ginsenoside Rh2 partially reduces changes in the expression levels of IL-1β and IL-18 in AMI rats. (g) Treatment with ginsenoside Rh2 partially recovers mRNA levels of IL-1β, IL-18, caspase-1, NLRP3, ASC and GSDND when compared with levels in AMI rats. *P<.05. (h) Treatment with ginsenoside Rh2 partially recovers protein levels of cleaved caspase-1, NLRP3, ASC, and GSDND when compared with levels in AMI rats. *P<.05. AMI, acute myocardial infarction; ASC, apoptosis-associated speck-like protein containing a caspase recruitment domain; IL-1β, interleukin-1β; IL-8, interleukin-8; GSDND, gasdermin D; LDH, lactate dehydrogenase; MDA, malondialdehyde; NLRP3, nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3.
Figure 1. Treatment with ginsenoside Rh2 alleviates AMI in rats.

Ginsenoside Rh2 reduced H/R-induced cardiomyocyte injury by regulating pyroptosis

We further examined the effects of ginsenoside Rh2 on H/R-induced cardiomyocyte injury. We found that the levels of MDA, LDH, IL-1β, and IL-18 were increased after H/R induction in H9c2 cells; however, treatment with ginsenoside Rh2 could partially reduce these levels (). Additionally, mRNA leveles of IL-1β, IL-18, caspase-1, NLRP3, ASC and GSDND were increased in H/R-induced H9c2 cells, which were partially reduced following ginsenoside Rh2 treatment (); And protein levels of cleaved caspase-1, NLRP3, ASC, and GSDND were elevated in H/R-induced H9c2 cells, which were partially reduced following ginsenoside Rh2 treatment ().

Figure 2. Treatment of ginsenoside Rh2 alleviates H/R-induced cell damage in H9c2 cells by regulating pyroptosis.

(a) and (b) Increased levels of MDA and LDH in H/R-induced H9c2 cells are partially reduced following treatment with ginsenoside Rh2. (c) Changes in expression levels of IL-1β and IL-18 in H9c2 with H/R-induced cells are partially reduced following treatment with ginsenoside Rh2. (d) Treatment with ginsenoside Rh2 partially recovers mRNA levels of IL-1β, IL-18, caspase-1, NLRP3, ASC and GSDND when compared with those in H/R-induced H9c2 cells. *P<.05. (e) Treatment with ginsenoside Rh2 partially recovers levels of cleaved Caspase-1, NLRP3, ASC, and GSDND protein when compared with those in H/R-induced H9c2 cells. *P<.05. ASC, apoptosis-associated speck-like protein containing a caspase recruitment domain; H/R, hypoxia/reoxygenation; IL-1β, interleukin-1β; IL-8, interleukin-8; GSDND, gasdermin D; LDH, lactate dehydrogenase; MDA, malondialdehyde; NLRP3, nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3.
Figure 2. Treatment of ginsenoside Rh2 alleviates H/R-induced cell damage in H9c2 cells by regulating pyroptosis.

Ginsenoside Rh2 regulated the PI3K/AKT pathway

To further investigate the possible molecular mechanisms underlying the ginsenoside Rh2-mediated inhibition of pyroptosis, we evaluated the activation status of the PI3K/AKT pathway. The results revealed that levels of p-PI3K and p-Akt were decreased, while these protein levels were partially increased following treatment with ginsenoside Rh2 ().

Figure 3. Ginsenoside Rh2 regulates the PI3K/AKT signaling pathway.

(a) Levels of p-PI3K and p-AKT in H9c2 cells decrease after H/R induction, but the levels are partially rescued by ginsenoside Rh2 treatment. **P<.01, ***P<.001. H/R, hypoxia/reoxygenation; PI3K/AKT, phosphoinositide 3-kinase/protein kinase B.
Figure 3. Ginsenoside Rh2 regulates the PI3K/AKT signaling pathway.

Discussion

It has been established that ginsenoside Rh2 plays an important role in regulating cardiovascular, central nervous, immune, and endocrine systems, affording beneficial effects in improving myocardial ischemia, as well as anti-allergy, anti-tumor, anti-depression, and anti-inflammatory effects (Citation20,Citation21). Ginsenoside Rh2 can significantly inhibit the production of reactive oxygen species in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages, indicating that ginsenoside Rh2 exerts beneficial inhibitory effects on the inflammatory response induced by bacterial endotoxins (Citation22). In addition, ginsenoside Rh2 can significantly reduce IL-6, tumor necrosis factor-α, and IL-1β released by RAW264.7 macrophages and promote the production of IL-10, which significantly inhibits LPS-induced macrophage inflammation (Citation21). Pyroptosis differs from apoptosis and necrosis, and inflammation often accompanies pyroptosis. GSDND is the main executor of pyroptosis and relies on the stimulation of inflammation-related caspases (Citation23,Citation24).

Pyroptosis is reportedly involved in the development of MI (Citation9). Pyroptosis of cardiomyocytes can promote the activation of caspase 1, maturation and release of IL-1β and IL-18, and enhance the inflammatory response (Citation25,Citation26). In the present study, cardiomyocyte pyroptosis increased cardiomyocyte necrosis and released MDA, LDH, IL-1β, and IL-18; however, treatment with ginsenoside Rh2 reversed these effects in vivo. Moreover, NLRP3 has been widely recognized as a major factor in pyroptosis induction (Citation27). Once NLRP3 is activated, it promotes the activation of other pyrogenic factors and subsequently induces pyroptosis (Citation28,Citation29). In the present study, NLRP3, ASC, cleaved caspase-1, and GSDND (which play an effector role in pyroptosis) exhibited increased protein expression levels in AMI. This finding indicates that AMI can induce an inflammatory response and pyroptosis. However, treatment with ginsenoside Rh2 could alleviate the high levels of pyroptosis-related protein expression. Accordingly, these results indicate that ginsenoside Rh2 treatment can relieve the AMI-induced inflammatory response and pyroptosis of cardiomyocytes, thereby suggesting the cardioprotective effect of ginsenoside Rh2 against inflammation and pyroptosis.

Studies have shown that Ginsenoside Rh2 participates in AMI by affecting pyroptosis of cells at the in vivo level (Citation15). However, it is still necessary to further understand the mechanism of Ginsenoside Rh2 in alleviating AMI by affecting pyroptosis. To the best of our knowledge, this is the first report regarding the effect of ginsenoside Rh2 on ameliorating AMI by inhibiting cardiomyocyte pyroptosis at the cellular level, and a possible molecular pathway was also elucidated. Herein, treatment with ginsenoside Rh2 significantly alleviated AMI and reduced pyroptosis-related factors in H/R-induced cardiomyocytes, suggesting that ginsenoside Rh2 exerts a cardioprotective effect by reversing cardiomyocyte pyroptosis during AMI. It is well known that PI3K and its downstream target, the serine/threonine kinase AKT, belong to a conserved family of signal transduction enzymes (Citation18,Citation30). The PI3K/AKT pathway is considered an important endogenous mechanism that promotes the survival of ischemic myocardial cells (Citation31). In the present study, treatment with ginsenoside Rh2 upregulated the expression of p-PI3K and p-Akt and activated the PI3K/AKT pathway. Accordingly, these data suggest that ginsenoside Rh2 may alleviate pyroptosis and AMI by activating the PI3K/AKT pathway. However, the detailed molecular mechanisms underlying this pathway warrant further investigation.

Conclusions

In conclusion, our current study demonstrated that ginsenoside Rh2 regulates pyroptosis in cardiomyocytes, alleviating AMI in vivo and in vitro. These findings could aid in developing new approaches for treating AMI.

Data sharing statement

The data sets used and analyzed during the current study are available from the corresponding author on reasonable request.

Ethics approval

The experimental and animal care procedures were approved by the Affiliated Hospital of Zunyi Medical University (Approval Number. ZMU21-2207-032) and conformed to the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

The author(s) reported there is no funding associated with the work featured in this article.

References

  • Zhang Y, Jiao L, Sun L, Li Y, Gao Y, Xu C, Shao Y, Li M, Li C, Lu Y, et al. LncRNA ZFAS1 as a SERCA2a inhibitor to cause intracellular Ca(2+) overload and contractile dysfunction in a mouse model of myocardial infarction. Circ Res. 2018;122(10):1354–7. doi:10.1161/CIRCRESAHA.117.312117.
  • Lu Y, Zhang Y, Shan H, Pan Z, Li X, Li B, Xu C, Zhang B, Zhang F, Dong D, et al. MicroRNA-1 downregulation by propranolol in a rat model of myocardial infarction: a new mechanism for ischaemic cardioprotection. Cardiovasc Res. 2009;84(3):434–41. doi:10.1093/cvr/cvp232.
  • Ibáñez B, Heusch G, Ovize M, Van de Werf F. Evolving therapies for myocardial ischemia/reperfusion injury. J Am Coll Cardiol. 2015;65(14):1454–71. doi:10.1016/j.jacc.2015.02.032.
  • Frangogiannis NG, Rosenzweig A. Regulation of the inflammatory response in cardiac repair. Circ Res. 2012;110(1):159–73. doi:10.1161/CIRCRESAHA.111.243162.
  • Daskalopoulos EP, Hermans KC, Blankesteijn WM. Cardiac (myo)fibroblast: novel strategies for its targeting following myocardial infarction. Curr Pharm Des. 2014;20(12):1987–2002. doi:10.2174/13816128113199990452.
  • Zychlinsky A, Prevost MC, Sansonetti PJ. Shigella flexneri induces apoptosis in infected macrophages. Nature. 1992;358(6382):167–69. doi:10.1038/358167a0.
  • Labbé K, Saleh M. Cell death in the host response to infection. Cell Death Differ. 2008;15(9):1339–49. doi:10.1038/cdd.2008.91.
  • Zhang Y, Liu X, Bai X, Lin Y, Li Z, Fu J, Li M, Zhao T, Yang H, Xu R, et al. Melatonin prevents endothelial cell pyroptosis via regulation of long noncoding RNA MEG3/miR-223/NLRP3 axis. J Pineal Res. 2018;64(2):20. doi:10.1111/jpi.12449.
  • Li Z, Xu H, Liu X, Hong Y, Lou H, Liu H, Bai X, Wang L, Li X, Monayo SM, et al. GDF11 inhibits cardiomyocyte pyroptosis and exerts cardioprotection in acute myocardial infarction mice by upregulation of transcription factor HOXA3. Cell Death Disease. 2020;11(10):020–03120. doi:10.1038/s41419-020-03120-6.
  • Ma M, Liang SC, Diao KY, Wang Q, He Y. Mitofilin mitigates myocardial damage in acute myocardial infarction by regulating pyroptosis of cardiomyocytes. Front Cardiovasc Med. 2022;9(823591). doi:10.3389/fcvm.2022.823591.
  • Li X, Chu S, Lin M, Gao Y, Liu Y, Yang S, Zhou X, Zhang Y, Hu Y, Wang H, et al. Anticancer property of ginsenoside Rh2 from ginseng. Eur J Med Chem. 2020;203(112627):13. doi:10.1016/j.ejmech.2020.112627.
  • Vinoth Kumar R, Oh TW, Park YK. Anti-inflammatory effects of ginsenoside-Rh2 inhibits LPS-Induced activation of Microglia and overproduction of inflammatory mediators via modulation of TGF-β1/Smad pathway. Neurochem Res. 2016;41(5):951–57. doi:10.1007/s11064-015-1804-x.
  • Lo SH, Hsu CT, Niu HS, Niu CS, Cheng JT, Chen ZC. Ginsenoside Rh2 improves cardiac fibrosis via PPARδ–STAT3 signaling in type 1-like diabetic rats. Int J Mol Sci. 2017;18(7):1364. doi:10.3390/ijms18071364.
  • Qi Z, Yan Z, Wang Y, Ji N, Yang X, Zhang A, Li M, Xu F, Zhang J. Ginsenoside Rh2 inhibits NLRP3 inflammasome activation and improves exosomes to alleviate hypoxia-induced myocardial injury. Front Immunol. 2022;13(883946). doi:10.3389/fimmu.2022.883946.
  • Song W, Dai B, Dai Y, Li W. Influence of gnsenoside Rh2 on cardiomyocyte pyroptosis in rats with acute myocardial infarction. Evid Based Complement Alternat Med. 2022;12(5194523):1–7. doi:10.1155/2022/5194523.
  • Qin W, Zhang L, Li Z, Xiao D, Zhang Y, Yang H, Zhang H, Xu C, Zhang Y. Metoprolol protects against myocardial infarction by inhibiting miR-1 expression in rats. J Pharm Pharmacol. 2020;72(1):76–83. doi:10.1111/jphp.13192.
  • Qi, Z, Yan, Z, Wang, Y et al. Integrative applications of network pharmacology and molecular docking: an herbal formula ameliorates H9c2 cells injury through pyroptosis. J Ginseng Res. 2023;47(2):228–236.
  • Xin G, Xu-Yong L, Shan H, Gang W, Zhen C, Ji-Jun L, Ping Y, Man-Hua C. SH2B1 protects cardiomyocytes from ischemia/reperfusion injury via the activation of the PI3K/AKT pathway. Int Immunopharmacol. 2020;83(105910):26. doi:10.1016/j.intimp.2019.105910.
  • Xu LJ, Chen RC, Ma XY, Zhu Y, Sun GB, Sun XB. Scutellarin protects against myocardial ischemia-reperfusion injury by suppressing NLRP3 inflammasome activation. Phytomedicine. 2020;68(153169):16. doi:10.1016/j.phymed.2020.153169.
  • Yang Y, Ren C, Zhang Y, Wu X. Ginseng: an nonnegligible natural remedy for healthy aging. Aging Dis. 2017;8(6):708–20. doi:10.14336/AD.2017.0707.
  • Bi WY, Fu BD, Shen HQ, Wei Q, Zhang C, Song Z, Qin QQ, Li HP, Lv S, Wu SC, et al. Sulfated derivative of 20(S)-ginsenoside Rh2 inhibits inflammatory cytokines through MAPKs and NF-kappa B pathways in LPS-induced RAW264.7 macrophages. Inflammation. 2012;35(5):1659–68. doi:10.1007/s10753-012-9482-1.
  • Park BG, Jung HJ, Cho YW, Lim HW, Lim CJ. Potentiation of antioxidative and anti-inflammatory properties of cultured wild ginseng root extract through probiotic fermentation. J Pharm Pharmacol. 2013;65(3):457–64. doi:10.1111/jphp.12004.
  • Zeng X, Zhu M, Liu X, Chen X, Yuan Y, Li L, Liu J, Lu Y, Cheng J, Chen Y. Oleic acid ameliorates palmitic acid induced hepatocellular lipotoxicity by inhibition of ER stress and pyroptosis. Nutr Metab. 2020;17(11):020–0434. doi:10.1186/s12986-020-0434-8.
  • He C, Zhao Y, Jiang X, Liang X, Yin L, Yin Z, Geng Y, Zhong Z, Song X, Zou Y, et al. Protective effect of Ketone musk on LPS/ATP-induced pyroptosis in J774A.1 cells through suppressing NLRP3/GSDMD pathway. Int Immunopharmacol. 2019;71:328–35.
  • Shi J, Gao W, Shao F. Pyroptosis: gasdermin-mediated programmed necrotic cell death. Trends Biochem Sci. 2017;42(4):245–54. doi:10.1016/j.tibs.2016.10.004.
  • Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol. 2009;7(2):99–109. doi:10.1038/nrmicro2070.
  • Schölwer I, Habib P, Voelz C, Rolfes L, Beyer C, Slowik A. NLRP3 depletion fails to mitigate inflammation but restores diminished phagocytosis in BV-2 cells after in vitro hypoxia. Mol Neurobiol. 2020;57(6):2588–99. doi:10.1007/s12035-020-01909-2.
  • Wu X, Zhang H, Qi W, Zhang Y, Li J, Li Z, Lin Y, Bai X, Liu X, Chen X, et al. Nicotine promotes atherosclerosis via ROS-NLRP3-mediated endothelial cell pyroptosis. Cell Death Disease. 2018;9(2):017–0257. doi:10.1038/s41419-017-0257-3.
  • Russo HM, Rathkey J, Boyd-Tressler A, Katsnelson MA, Abbott DW, Dubyak GR. Active caspase-1 induces plasma membrane pores that precede pyroptotic lysis and are blocked by lanthanides. J Immunol. 2016;197(4):1353–67. doi:10.4049/jimmunol.1600699.
  • Guo X, Jiang H, Chen J. RP105-PI3K-Akt axis: a potential therapeutic approach for ameliorating myocardial ischemia/reperfusion injury. Int J Cardiol. 2016 Mar 1;206:95–96. [accsessed 2016 Jan 8]. doi:10.1016/j.ijcard.2016.01.100.
  • Qin Q, Cui L, Zhou Z, Zhang Z, Wang Y, Zhou C. Inhibition of microRNA-141-3p reduces hypoxia-induced apoptosis in H9c2 rat cardiomyocytes by activating the RP105-dependent PI3K/AKT signaling Pathway. Med Sci Monit. 2019;25:7016–25. doi:10.12659/MSM.916361.