Abstract
Heart failure (HF) is a disease with high mortality and morbidity rate. Autophagy is critically implicated in HF progression. The current research was designed to investigate the function of Dioscin on oxidative stress, autophagy, and apoptosis in HF. In this study, doxorubicin (Dox) was employed to induce HF model and HL‐1 cell damage model. Echocardiography implied that Dioscin could dramatically relieve heart function in vivo. Western blotting determined that Dioscin treatment reversed the promotive effect of autophagy caused by Dox through modulating levels of key autophagy‐associated molecules, including Atg5 and Beclin1. Dioscin also impaired apoptosis by regulating apoptosis‐related protein, including Bcl‐2 and cleaved caspase‐3 following Dox treatment in vivo and in vitro. Furthermore, the impacts of Dioscin were mediated by upregulation of PDK1‐mediated Akt/mTOR signaling. The mTOR inhibitor (rapamycin) could counteract the therapeutic impact of Dioscin in vitro. Taken together, Dioscin could relieve cardiac function through blocking apoptosis and autophagy by activating the PDK1‐elicited Akt/mTOR pathway.
Keywords: Akt/mTOR, Dioscin, doxorubicin, heart failure, PDK1
1. INTRODUCTION
Heart failure (HF) is a global medical problem and complex clinical syndrome, which is the final stage of a wide range of cardiovascular diseases characterized by impaired heart capacity to contract or relax, and the leading cause of cardiac disease‐associated death. 1 , 2 It was demonstrated that HF was closely related to cardiac hypertrophy, dilated cardiomyopathy, hypertrophic cardiomyopathy, age, obesity, diabetes, and hypertension. 3 , 4 Even though the availability of effective treatments aimed at ameliorating HF, the 5‐year death rate from the disease is still close to 50%. 5 In the pathological process of HF, apoptosis leads to the reduction of cardiomyocytes and the decrease of myocardial contractility, thus boosting the occurrence and development of HF, confirming the vital function of apoptosis in HF. 6 Autophagy is a conserved system in eukaryotic cells to recycle damaged organelles and proteins, which plays a crucial function in maintaining cell homeostasis and survival. However, overactivation of autophagy has also been shown to be harmful. For instance, stimulated autophagy by myocardial reperfusion can lead to cardiac injury. 7 Similarly, autophagy elicited by doxorubicin (Dox) treatment in myocardium promoted the cardiotoxicity of Dox. 8 Thus, autophagy inhibition could be a potential strategy for the treatment of Dox‐triggered cardiomyopathy. Notably, Dox was widely used to produce animal and cellular models of HF. 9 , 10 Therefore, a thorough understanding of the mechanism of cardiomyocyte apoptosis and autophagy in Dox‐induced HF is imperative to develop effective treatment methods and evaluate the prognosis of HF patients.
Phosphoinositide dependent protein kinase‐1 (PDK1) is a serine/threonine kinase belonging to the AGC kinase family. It was reported that PDK1 plays a fundamental function in the PI3K‐PDK1‐Akt pathway. 11 Moreover, PI3K activation resulted in the synthesis of phosphatidylinositol‐3,4,5‐triphosphate. 12 Afterward, PDK1 and Akt are recruited to the plasma membrane, and then the membrane‐bound PDK1 phosphorylates Akt at Thr308, resulting in activating Akt, which was involved in cardiovascular pathological processes such as atherosclerosis, myocardial hypertrophy, and vascular remodeling. 13 , 14 Recently, Zhu et al. manifested that depletion of Mirt2 aggravated ischemia‐triggered myocardial infarction and cardiomyocyte apoptosis through reducing PDK1. 15 Gao et al. elucidated that HTR2A enhanced phosphorylations of PDK1 to activate AKT‐mTOR signaling and promoted cardiac hypertrophy. 16 Furthermore, it was revealed that deficiency of PDK1 in cardiac muscle results in HF and increased sensitivity to hypoxia. 17 This research explored the impact of PDK1 on Dox‐induced HF.
Dioscin is a kind of steroid sapogenin synthesized naturally by plants, belonging to spirosterol class. As an essential raw material for the synthesis of steroid hormones and steroidal contraceptives, Dioscin was commonly employed for the production of pregnenolone, progesterone, and cortisol drugs. 18 In the past few decades, pharmacological tests have implied that Dioscin has the effects of antitumor, modulating blood lipids, preventing platelets aggregation, and accelerating bile secretion; thus, it is mainly employed to treat cardiovascular disease, encephalitis, and cancers. 19 For instance, Dioscin suppressed coronary heart disease via decreasing oxidative stress by modulating p38 MAPK signaling. 20 What is more, Dioscin was reported to treat myocardial infarction by promoting angiogenesis. 21 However, whether Dioscin protected the heart from damage remains unclear.
In this work, the effects of Dioscin on autophagy and PDK1/Akt/mTOR signaling were explored by in vivo and in vitro experiments. This research will offer a prospective therapeutic target and prognostic indicator for HF patients.
2. MATERIALS AND METHODS
2.1. Animals and HF model establishment
All of the animal experimental protocols were subject to approval by Jiangsu Province Hospital of Chinese Medicine (Approval No.: NU(2020) 3A‐152). Sixteen male C57/BL6 mice (8 weeks old) were gained from the Laboratory Animal (Nanjing).
HF model in mice was constructed using Dox. 22 Dox (cat. no. 25316‐40‐9; MilliporeSigma) was dissolved in saline to generate a stock solution of 100 μM and was diluted to a final concentration of 2 μM for all experiments unless otherwise specified. Briefly, following 7 days acclimation, mice were randomly divided into the Control (control, n = 8) and Dox group (HF group, n = 8). Mice in the HF group were intraperitoneally of 2.5 mg/kg injected with Dox six times in 2 weeks, and then were treated with 40 mg/kg of Dioscin for seven consecutive days by oral gavage. 23 Dioscin (purity >98%) was purchased from Spring & Autumn Biological Engineering Co., Ltd. (Nanjing, China) All mice in control groups were treated with 0.5% CMC‐Na for seven consecutive days. Finally, the electrocardiograms of mice were detected before the animals were sacrificed. Mice were sacrificed under anesthesia to relieve pain. Heart tissues were gathered at −80°C for further analysis.
2.2. Echocardiographic assessment
Mice were anesthetized with 1% of the pentobarbital sodium with a dose of 50 mg/kg. The left ventricular systolic pressure (LVSP), left ventricular end‐diastolic pressure (LVEDP), and ±dp/dt max parameters of mice were assessed by the hemodynamic method.
2.3. Cell culture, transfection, and in vitro experimental design
Mouse myocardial cell line (HL‐1) was purchased from American Type Culture Collection (ATCC, Manassas, USA), cultured in the DMEM medium containing 10% FBS (Hyclone, USA), 100 μg/mL of streptomycin, and 100 U/mL of penicillin (Hyclone, USA), and incubated at 37°C with 5% CO2. When the cells were grown to 70% confluence, the knockdown vector of PDK1 (shPDK1) and control vectors (shNC) were transfected using Lipofectamine 2000 (Invitrogen, USA). Cells were harvested after 48 h transfection for further experiments. To examine the role of Dioscin in myocardial injury cell model, HL‐1 cells were pretreated with 100 ng/mL Dioscin for 24 h before challenged with Dox (5 μM) for 24 h. 23
2.4. Histopathology analysis
For HE staining, the heart tissues were fixed with 4% paraformaldehyde (Beyotime Institute of Biotechnology) and then cut into 0.4 μm slices after staining with H&E. The pathological alterations were observed under a microscope. For Immunohistochemistry, the heart sections were gathered and incubated with the antibody according to previously reported recommendations. 24
2.5. Biochemical analysis
The levels of the lipid peroxidation biomarker malondialdehyde (MDA) and the activity of antioxidant enzymes superoxide dismutase (SOD) were determined using an appropriate commercial kit (Bio‐Diagnostics Co, Egypt).
2.6. Western blotting
Total proteins were exacted from mouse heart tissues and myocardial cells with RIPA buffer (Beyotime). Subsequently, the proteins were separated by SDS‐PAGE, transferred onto PVDF membranes, and then blocked with 5% nonfat milk at 37°C for 1 h. Thereafter, the membranes were probed with primary antibodies p‐PDK1Ser241 (1:500, ab131098, abcam), PDK1 (1:1000, ab202468, abcam), cleaved caspase 3 (1:1000, ab32042, Abcam), Bcl‐2 (1:1000, ab182858, Abcam), Beclin1 (1:1000, ab207612, Abcam), Atg5 (1:100, ab109490, Abcam), p‐Akt (1:500, ab38449, Abcam), p‐mTOR (1:1000, ab109268, Abcam), Akt (1:500, ab8805, Abcam), mTOR (1:500, ab32028, Abcam) and GAPDH (1:1000, 2803, CST) at 4°C overnight and secondary antibodies for nearly 2 h. Protein bands were visualized with ECL kit (Pierce) and quantified by Image J software.
2.7. Flow cytometry
HL‐1 cells were rinsed with cold PBS and then re‐suspended in Annexin V Binding Buffer. Next, HL‐1 cells were added with FITC‐Annexin V and PI (BD Pharmingen). Thereafter, data were assessed via flow cytometry with FACScalibur flow cytometer (Becton Dickinson).
2.8. Statistical analysis
The GraphPad Prism 7 software was employed for statistical analysis. The data were expressed as mean ± SD from at least three times of experiments independently. One‐way ANOVA followed by Tukey's post hoc test was used to compare statistical differences among various groups. p < 0.05 was deemed significant in statistics.
3. RESULTS
3.1. The effect of Dioscin on the cardiac function and structural changes in Dox‐induced HF mice
First, echocardiography revealed that HF model in mice was established successfully, evidenced by the obvious reduction in the level of LVSP and enhancement in the level of LVEDP in Dox‐induced HF mice compared with the control mice. Then, after Dioscin treatment, LVSP level was heightened, and LVEDP was reduced (Figure 1A,B). HE staining displayed that the left ventricular myocardial cells in the control group were arranged orderly, while those in the HF group were arranged loosely. The pathological changes were counteracted by Dioscin introduction (Figure 1C). These results expounded that Dioscin alleviated Dox‐induced cardiac damage in HF mice.
FIGURE 1.

Effect of Dioscin on the cardiac function and structural changes in DOX‐induced HF mice. (A,B) Echocardiographic analysis showed the effect of Dioscin on LVSP and LVEDP values. (C) HE exhibited that cardiomyocyte architecture in mice treated with Dox or Dox + Dioscin. *p < 0.05; **p < 0.01. N = 3 per group.
3.2. Dioscin suppresses Dox‐induced apoptosis and excessive autophagy in HF mice model
The levels of key molecules in the apoptotic pathway including Bcl‐2 and Cleaved‐caspase‐3 were examined via western blotting. As depicted in Figure 2A, the levels of pro‐apoptotic protein (cleaved caspase 3) were augmented, whereas the anti‐apoptotic protein (Bcl‐2) was lessened in Dox‐elicited HF mice, suggesting that the apoptotic signaling was activated. However, treatment with Dioscin reversed the levels of these proteins toward normal levels, suggesting that Dioscin had an anti‐apoptotic effect on HF mice model. Accumulating evidence has implied that autophagy dysfunction is the main cause of cardiac fibrosis. 25 To further elucidated the role of autophagy in HF mice model, protein expression of classical autophagic markers including Beclin1 and Atg5 was measured by Western blotting. Results insinuated that protein levels of Beclin1 and Atg5 were elevated in Dox‐elicited cardiac tissue. Meanwhile, Dioscin treatment downregulated the levels of Beclin1 and Atg5, implying that Dioscin could attenuate autophagy (Figure 2B). These results expounded that autophagy in the Dioscin‐treated mice was suppressed compared with that in Dox administration mice, which thereby suggested that autophagy could be a key regulatory mechanism in the protective impact of Dioscin.
FIGURE 2.

Dioscin suppresses Dox‐induced apoptosis and excessive autophagy in HF mice model. (A) Western blotting detected the level of Bcl‐2, cleaved caspase 3 in mice treated with Dox or Dox + Dioscin. (B) Protein expression of Beclin1 and Atg5 were analyzed. *p < 0.05; **p < 0.01. N = 3 per group.
3.3. Dioscin protects HL‐1 cells from injuries caused by Dox
To further uncover the potential target of Dioscin on HF, the Dox‐stimulated HL‐1 cell model was established. Flow cytometry assay showed that the apoptosis augmented by Dox stimulation, which was substantially lessened by Dioscin (Figure 3A). Besides, Dioscin abrogated the enhancement of cleaved caspase 3 levels and reduction of Bcl‐2 induced by Dox (Figure 3B). Detection of MDA and SOD levels in HL‐1 cells demonstrated that Dioscin had an anti‐oxidant effect in vitro (Figure 3C,D). Taken together, Dioscin ameliorated Dox‐induced cell apoptosis and oxidative stress in HL‐1 cells.
FIGURE 3.

Dioscin protects HL‐1 cells from the injuries caused by Dox. HL‐1 cells were grouped into Control, Dox, and Dox + Dioscin groups. (A) Cell death was detected using flow cytometry assay. (B) Protein levels of cleaved caspase 3 and Bcl‐2. (C,D) Levels of MDA and SOD. *p < 0.05; **p < 0.01. N = 3 per group.
3.4. Dioscin activates Akt/mTOR signaling via PDK1
Subsequently, the mechanism underlying the effect of Dioscin was studied. It was displayed that the protein levels of PDK1, p‐Akt, and p‐mTOR were decreased following Dox treatment, which was neutralized by Dioscin. To confirm whether Dioscin regulated HF progression via the PDK1/Akt/mTOR pathway, PDK1 was silenced in HL‐1 cells. Western blotting showed that PDK1 deficiency reversed the promotion of Dioscin on p‐PDK1 (Ser241), p‐Akt, and p‐mTOR levels in Dox‐induced HL‐1 cells, suggesting that PDK1 activated the Akt/mTOR signaling (Figure 4A). Next, rapamycin (inhibitor of mTOR) was introduced. Similarly, the p‐Akt, and p‐mTOR protein level was decreased in Dox + Dioscin + rapamycin group compared with that in Dox + Dioscin group. In the meantime, the total Akt and mTOR protein levels were not affected. These results implicated that Dioscin could upregulate the activity of PDK1/Akt/mTOR pathway in Dox‐elicited HL‐1 cells.
FIGURE 4.

Dioscin activates Akt/mTOR signaling via PDK1. (A) The protein expression of p‐PDK1 (Ser241), p‐Akt, p‐mTOR, PDK1, Akt, and mTOR in HL‐1 cells in Control, Dox, Dox + Dioscin, and Dox + Dioscin + shPDK1 and Dox + Dioscin + rapamycin groups was detected. *p < 0.05. N = 3 per group.
3.5. Dioscin prevents cell apoptosis and autophagy in vitro through PDK1‐mediated Akt/mTOR signaling
It was elaborated that PDK1, as a target protein, played an essential function via the Akt/mTOR pathway in the processes of autophagy and apoptosis. 26 The biological role of PDK1‐mediated Akt/mTOR signaling in HF progression was evaluated. As exhibited in Figure 5A, the suppressive effect of Dioscin on the apoptosis of HL‐1 cells under Dox stimulation was abated by PDK1 deficiency or rapamycin. Consistently, PDK1 knockdown attenuated the effects of Dioscin on the levels of cleaved caspase 3 and Bcl‐2, MDA and SOD. Similarly, inactivation of the Akt/mTOR signaling has same effects as PDK1 depletion (Figure 5B–D). Furthermore, western blotting showed that rapamycin or shPDK1 abrogated the effects of Dioscin on Beclin1 and Atg5 (Figure 5E). In sum, Dioscin prevented HL‐1 cells from Dox‐induced damages via activating the PDK1/Akt/mTOR pathway.
FIGURE 5.

Dioscin prevents cell apoptosis and provokes autophagy in vitro through PDK1‐mediated Akt/mTOR signaling. Cells were divided into Dox + Dioscin, Dox + Dioscin + shPDK1, and Dox + Dioscin + rapamycin groups. (A) The apoptosis of designated cells was detected. (B–D) The levels of cleaved caspase 3, Bcl‐2, MDA, and SOD levels. (E) Protein expression of Beclin1 and Atg5. *p < 0.05; **p < 0.01. N = 3 per group.
4. DISCUSSION
The present study provided evidence that Dioscin ameliorates Dox‐induced HF via inhibiting autophagy and apoptosis via in vivo and in vitro studies. It has been widely recognized that estradiol decreases the risk for cardiovascular disease, but estrogen replacement therapy is limited for its side effects. 27 Dioscin, a natural steroid saponin isolated from the root bark of wild dioscorea nipponica, is currently widely employed for cardiovascular disease treatment. 28 It was implied by Kong that Dioscin alleviated hypoxic‐caused cardiac dysfunction in myocardial infarction via elevating the level of lncRNA MANTIS. 29 Dioscin improved myocardial infarction damage through adjusting BMP4/NOX1‐mediated inflammation. 30 In addition, Dox was reported as an effective and commonly used to treat hematological and solid tumors. 31 However, Dox can lead to multiple‐organ toxicity in patients. As an anticancer agent, Dox can lead to long‐term dose‐dependent cardiotoxicity and HF. 32 This research elaborated that Dioscin protected heart function in Dox‐induced HF mice model and HL‐1 cell model, evidenced by improved LVSP and reduced LVEDP. The findings of this study indicated that Dioscin could suppress Dox‐induced HF and provided a new therapeutic method for HF.
Next, we further excavated the influences and mechanisms of Dioscin on autophagy. It was insinuated that autophagy is a dynamic process that participated in the concerted action of autophagy‐related genes, including Beclin‐1, Atg5, and LC3. 33 Multiple studies proposed that disordered and reduced autophagy was involved with HF progression. Moxibustion impaired chronic HF via repressing autophagy by elevating mTOR expression. 34 Rutin relieved Dox‐induced HF by reducing excessive autophagy and apoptosis. 35 Previous studies disclosed that Dox‐triggered cardiotoxicity was implicated in the dysregulation of autophagy. Moreover, Dox administration promoted myocardial autophagy, which was closely related to damage to cardiac function. 36 Excessive autophagy can induce cell death, which is called autophagic cell death. Several researches have elaborated that the suppression of Dox‐triggered excessive autophagy has vital beneficial impacts on cardiac damage in vivo. 37 Herein, autophagy was activated by Dox in cardiac tissue, and Dioscin may provide protection by restraining excessive autophagy. Further, it was implied that Dioscin suppressed Dox‐induced apoptosis and excessive autophagy in HF mice model, and ameliorated Dox‐induced cell apoptosis and oxidative stress in HL‐1 cells. These findings unveiled that Dioscin protected against myocardial cell damage by repressing apoptosis and autophagy.
Subsequently, we further verified the mechanism of Dioscin in regulating autophagy and PDK1/Akt/mTOR signaling pathway in HF progression. It was illustrated that PDK1 was closely implicated in myocardial cell injury via the Akt/mTOR pathway in myocardial cell injury. To cite an instance, PDK1 has been well established to activate Akt/mTOR signaling, and deficiency of PDK1 triggered severe cardiomyopathy and aberrant electrophysiology. 38 Moreover, LDHA might have effects on ischemia‐reperfusion injury and accelerate cardiomyocyte proliferation by affecting the expression of PDK1, Akt, and mTOR. 39 Herein, it was demonstrated that Dox does not inhibited the p‐PDK1/PDK1 ratio, but directly inhibited the PDK1 protein. Moreover, Dioscin treatment partially reversed the PDK1 level decreased by Dox. Furthermore, rapamycin or shPDK1 reversed the activation of Dioscin on PDK1 and Akt/mTOR signaling in Dox‐elicited HL‐1 cells. Besides, the effects of Dioscin on Dox‐stimulated HL‐1 cell apoptosis, oxidative stress, and autophagy‐associated proteins Beclin1 and Atg5 were offset after co‐incubation with rapamycin or PDK1 silence, indicating that the mechanism of Dioscin in HF is mediated by PDK1/Akt/mTOR signaling.
In conclusion, the function and mechanism of Dioscin have been investigated, and our work demonstrated Dioscin exerted cardioprotective effect through inhibiting Dox‐induced apoptosis and excessive autophagy via regulating the PDK1/Akt/mTOR pathway. These discoveries provide a theoretical basis for studying the pharmacological mechanism of Dioscin and a novel therapeutic strategy for HF.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest.
Yuan L, Ji H‐G, Yan X‐J, Liu M, Ding Y‐H, Chen X‐H. Dioscin ameliorates doxorubicin‐induced heart failure via inhibiting autophagy and apoptosis by controlling the PDK1‐mediated Akt/mTOR signaling pathway. Kaohsiung J Med Sci. 2023;39(10):1022–1029. 10.1002/kjm2.12740
Ling Yuan and Hai‐Gang Ji contributed equally to this research and should be considered as co‐first author.
REFERENCES
- 1. McMurray JJ, Adamopoulos S, Anker SD, Auricchio A, Böhm M, Dickstein K, et al. ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012. Turk Kardiyol Dern Ars. 2012;40(Suppl 3):77–137. [PubMed] [Google Scholar]
- 2. Tsutsui H. Recent advances in the pharmacological therapy of chronic heart failure: evidence and guidelines. Pharmacol Ther. 2022;238:108185. [DOI] [PubMed] [Google Scholar]
- 3. Piek A, de Boer RA, Silljé HH. The fibrosis‐cell death axis in heart failure. Heart Fail Rev. 2016;21:199–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Mosterd A, Hoes AW. Clinical epidemiology of heart failure. Heart. 2007;93:1137–1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, et al. Heart disease and stroke statistics‐2017 update: a report from the American Heart Association. Circulation. 2017;135:e146–e603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Shi B, Huang Y, Ni J, Chen J, Wei J, Gao H, et al. Qi Dan Li Xin pill improves chronic heart failure by regulating mTOR/p70S6k‐mediated autophagy and inhibiting apoptosis. Sci Rep. 2020;10:6105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Long L, Yu Z, Chen S, Wu J, Liu Y, Peng J, et al. Pretreatment of Huoxue Jiedu formula ameliorates myocardial Ischaemia/reperfusion injury by decreasing autophagy via activation of the PI3K/AKT/mTOR pathway. Front Pharmacol. 2021;12:608790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Li S, Wang W, Niu T, Wang H, Li B, Shao L, et al. Nrf2 deficiency exaggerates doxorubicin‐induced cardiotoxicity and cardiac dysfunction. Oxid Med Cell Longev. 2014;2014:748524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Lam PY, Kutchukian P, Anand R, Imbriglio J, Andrews C, Padilla H, et al. Cyp1 inhibition prevents doxorubicin‐induced cardiomyopathy in a zebrafish heart‐failure model. Chembiochem. 2020;21:1905–1910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Xu S, Wang Y, Yu M, Wang D, Liang Y, Chen Y, et al. LongShengZhi capsule inhibits doxorubicin‐induced heart failure by anti‐oxidative stress. Biomed Pharmacother. 2020;123:109803. [DOI] [PubMed] [Google Scholar]
- 11. Choi JH, Yang YR, Lee SK, Kim SH, Kim YH, Cha JY, et al. Potential inhibition of PDK1/Akt signaling by phenothiazines suppresses cancer cell proliferation and survival. Ann N Y Acad Sci. 2008;1138:393–403. [DOI] [PubMed] [Google Scholar]
- 12. Toker A, Newton AC. Cellular signaling: pivoting around PDK‐1. Cell. 2000;103:185–188. [DOI] [PubMed] [Google Scholar]
- 13. Abeyrathna P, Su Y. The critical role of Akt in cardiovascular function. Vascul Pharmacol. 2015;74:38–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Moon G, Kim J, Min Y, Wi SM, Shim JH, Chun E, et al. Phosphoinositide‐dependent kinase‐1 inhibits TRAF6 ubiquitination by interrupting the formation of TAK1‐TAB2 complex in TLR4 signaling. Cell Signal. 2015;27:2524–2533. [DOI] [PubMed] [Google Scholar]
- 15. Zhu F, Li Q, Li J, Li B, Li D. Long noncoding Mirt2 reduces apoptosis to alleviate myocardial infarction through regulation of the miR‐764/PDK1 axis. Lab Invest. 2021;101:165–176. [DOI] [PubMed] [Google Scholar]
- 16. Gao W, Guo N, Zhao S. HTR2A promotes the development of cardiac hypertrophy by activating PI3K‐PDK1‐AKT‐mTOR signaling. Cell Stress Chaperones. 2020;25:899–908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Mora A, Davies AM, Bertrand L, Sharif I, Budas GR, Jovanović S, et al. Deficiency of PDK1 in cardiac muscle results in heart failure and increased sensitivity to hypoxia. EMBO J. 2003;22:4666–4676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Zeng H, Yang L, Zhang X, Chen Y, Cai J. Dioscin prevents LPS‐induced acute lung injury through inhibiting the TLR4/MyD88 signaling pathway via upregulation of HSP70. Mol Med Rep. 2018;17:6752–6758. [DOI] [PubMed] [Google Scholar]
- 19. Gu L, Tao X, Xu Y, Han X, Qi Y, Xu L, et al. Dioscin alleviates BDL‐ and DMN‐induced hepatic fibrosis via Sirt1/Nrf2‐mediated inhibition of p38 MAPK pathway. Toxicol Appl Pharmacol. 2016;292:19–29. [DOI] [PubMed] [Google Scholar]
- 20. Yang B, Xu B, Zhao H, Wang YB, Zhang J, Li CW, et al. Dioscin protects against coronary heart disease by reducing oxidative stress and inflammation via Sirt1/Nrf2 and p38 MAPK pathways. Mol Med Rep. 2018;18:973–980. [DOI] [PubMed] [Google Scholar]
- 21. Shen T, Lyu D, Zhang M, Shang H, Lu Q. Dioscin alleviates cardiac dysfunction in acute myocardial infarction via rescuing mitochondrial malfunction. Front Cardiovasc Med. 2022;9:783426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Chen L, Yan KP, Liu XC, Wang W, Li C, Li M, et al. Valsartan regulates TGF‐β/Smads and TGF‐β/p38 pathways through lncRNA CHRF to improve doxorubicin‐induced heart failure. Arch Pharm Res. 2018;41:101–109. [DOI] [PubMed] [Google Scholar]
- 23. Zhao L, Tao X, Qi Y, Xu L, Yin L, Peng J. Protective effect of dioscin against doxorubicin‐induced cardiotoxicity via adjusting microRNA‐140‐5p‐mediated myocardial oxidative stress. Redox Biol. 2018;16:189–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Brooks HL, Lindsey ML. Guidelines for authors and reviewers on antibody use in physiology studies. 2018;314:H724–H732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Lee SY, Lee AR, Choi JW, Lee CR, Cho KH, Lee JH, et al. IL‐17 induces autophagy dysfunction to promote inflammatory cell death and fibrosis in keloid fibroblasts via the STAT3 and HIF‐1α dependent signaling pathways. Front Immunol. 2022;13:888719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Meng F, Zhang Y, Li X, Wang J, Wang Z. Clinical significance of miR‐138 in patients with malignant melanoma through targeting of PDK1 in the PI3K/AKT autophagy signaling pathway. Oncol Rep. 2017;38:1655–1662. [DOI] [PubMed] [Google Scholar]
- 27. Lan XF, Zhang XJ, Lin YN, Wang Q, Xu HJ, Zhou LN, et al. Estradiol regulates Txnip and prevents intermittent hypoxia‐induced vascular injury. Sci Rep. 2017;7:10318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Lyu D, Tian Q, Qian H, He C, Shen T, Xi J, et al. Dioscin attenuates myocardial ischemic/reperfusion‐induced cardiac dysfunction through suppression of reactive oxygen species. Oxid Med Cell Longev. 2021;2021:3766919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Kong C, Lyu D, He C, Li R, Lu Q. Dioscin elevates lncRNA MANTIS in therapeutic angiogenesis for heart diseases. Aging Cell. 2021;20:e13392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Zhang Z, Zhao X, Gao M, Xu L, Qi Y, Wang J, et al. Dioscin alleviates myocardial infarction injury via regulating BMP4/NOX1‐mediated oxidative stress and inflammation. Phytomedicine. 2022;103:154222. [DOI] [PubMed] [Google Scholar]
- 31. Cardinale D, Colombo A, Bacchiani G, Tedeschi I, Meroni CA, Veglia F, et al. Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation. 2015;131:1981–1988. [DOI] [PubMed] [Google Scholar]
- 32. QuanJun Y, GenJin Y, LiLi W, YongLong H, Yan H, Jie L, et al. Protective effects of dexrazoxane against doxorubicin‐induced cardiotoxicity: a metabolomic study. PloS One. 2017;12:e0169567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Shrestha A, Pun NT, Park PH. ZFP36L1 and AUF1 induction contribute to the suppression of inflammatory mediators expression by globular adiponectin via autophagy induction in macrophages. Biomol Ther. 2018;26:446–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Li Q, Wang W, Ma Q, Xia R, Gao B, Zhu G, et al. Moxibustion improves chronic heart failure by inhibiting autophagy and inflammation via upregulation of mTOR expression. Evid Based Complement Alternat Med. 2021;2021:6635876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Ma Y, Yang L, Ma J, Lu L, Wang X, Ren J, et al. Rutin attenuates doxorubicin‐induced cardiotoxicity via regulating autophagy and apoptosis. Biochim Biophys Acta Mol Basis Dis. 2017;1863:1904–1911. [DOI] [PubMed] [Google Scholar]
- 36. Bartlett JJ, Trivedi PC, Pulinilkunnil T. Autophagic dysregulation in doxorubicin cardiomyopathy. J Mol Cell Cardiol. 2017;104:1–8. [DOI] [PubMed] [Google Scholar]
- 37. Xu ZM, Li CB, Liu QL, Li P, Yang H. Ginsenoside Rg1 prevents doxorubicin‐induced cardiotoxicity through the inhibition of autophagy and endoplasmic reticulum stress in mice. Int J Mol Sci. 2018;19:3658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Qian XJ, Li XL, Xu X, Wang X, Feng QT, Yang CJ. α‐SMA‐Cre‐mediated excision of PDK1 reveals an essential role of PDK1 in regulating morphology of cardiomyocyte and tumor progression in tissue microenvironment. Pathol Biol. 2015;63:91–100. [DOI] [PubMed] [Google Scholar]
- 39. Du W, Qi X, Liu G, Zhao X, Yuan J, Wen J. Effects of target regulation of LDHA through the PDK1/Akt/mTOR pathway on myocardial apoptosis caused by ischemia/reperfusion injury in rats. Int J Clin Exp Med. 2017;10:14194–14202. [Google Scholar]
