Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Cinnamaldehyde alleviates doxorubicin-induced cardiotoxicity by decreasing oxidative stress and ferroptosis in cardiomyocytes

  • Meijiao Mao ,

    Contributed equally to this work with: Meijiao Mao, Wang Zheng

    Roles Conceptualization, Methodology, Project administration, Writing – original draft, Writing – review & editing

    amao1236@163.com (MM); lhzhna@163.com (NZ)

    Affiliation Department of Cardiology, Longhua Hospital, ShangHai University of Traditional Chinese Medicine, Shanghai, China

  • Wang Zheng ,

    Contributed equally to this work with: Meijiao Mao, Wang Zheng

    Roles Conceptualization, Formal analysis, Funding acquisition

    Affiliation Department of Cardiology, Longhua Hospital, ShangHai University of Traditional Chinese Medicine, Shanghai, China

  • Bin Deng,

    Roles Funding acquisition

    Affiliation Department of Cardiology, Longhua Hospital, ShangHai University of Traditional Chinese Medicine, Shanghai, China

  • Youhua Wang,

    Roles Funding acquisition, Methodology

    Affiliation Department of Cardiology, Longhua Hospital, ShangHai University of Traditional Chinese Medicine, Shanghai, China

  • Duan Zhou,

    Roles Project administration

    Affiliation Department of Cardiology, Longhua Hospital, ShangHai University of Traditional Chinese Medicine, Shanghai, China

  • Lin Shen,

    Roles Investigation

    Affiliation Department of Cardiology, Longhua Hospital, ShangHai University of Traditional Chinese Medicine, Shanghai, China

  • Wankang Niku,

    Roles Data curation

    Affiliation Department of Cardiology, Longhua Hospital, ShangHai University of Traditional Chinese Medicine, Shanghai, China

  • Na Zhang

    Roles Data curation, Supervision, Writing – review & editing

    amao1236@163.com (MM); lhzhna@163.com (NZ)

    Affiliation Department of Cardiology, Longhua Hospital, ShangHai University of Traditional Chinese Medicine, Shanghai, China

Abstract

Although doxorubicin (DOX) is an efficient chemotherapeutic drug for human tumors, severe cardiotoxicity restricts its clinical use. Cinnamaldehyde (CA), a bioactive component isolated from Cinnamonum cassia, possesses potent anti-oxidative and anti-apoptotic potentials. The major aim of this study was to evaluate the protective role of CA against DOX-induced cardiotoxicity. To this end, cardiomyocyte injury models were developed using DOX-treated H9c2 cells and DOX-treated rats, respectively. Herein, we found that CA treatment increased cardiomyocyte viability and attenuated DOX-induced cardiomyocyte death in vitro. CA further protected rats against DOX-induced cardiotoxicity, as indicated by elevated creatine kinase (CK) and lactate dehydrogenase (LDH) levels, myocardium injury, and myocardial fibrosis. CA alleviated DOX-induced myocardial oxidative stress by regulating reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione (GSH) levels. Mechanistically, CA markedly accelerated nuclear translocation of nuclear erythroid factor 2-related factor 2 (Nrf2) and increased heme oxygenase-1 (HO-1) expression. Consequently, CA decreased DOX-induced cardiomyocyte ferroptosis, while Erastin (a ferroptosis agonist) treatment destroyed the effect of CA on increasing cardiomyocyte viability. Taken together, the current results demonstrate that CA alleviates DOX-induced cardiotoxicity, providing a promising opportunity to increase the clinical application of DOX.

Introduction

Despite its proven efficacy in treating multiple types of tumors, the clinical application of DOX is restricted by its severe cardiotoxicity [1, 2]. Emerging evidence has demonstrated that DOX-induced myocardial damage is closely correlated with mitochondrial impairment [3], intracellular calcium homeostasis [4, 5], cardiomyocyte apoptosis [6], ferroptosis [7, 8], and oxidative stress [9, 10].

Oxidative stress is considered a vital cause of DOX-triggered myocardial damage [11]. Semiquinone, derived from DOX via one-electron reduction, can autoxidize by transferring an unpaired electron to O2 and thus generate a superoxide anions free radical (O2-) [11, 12]. O2- results in DNA damage, mitochondrial impairment, and subsequent cardiomyocyte injury [11]. Oxidative stress raises cardiomyocyte Ca2+ levels by influencing the sarcolemmal membrane to facilitate Ca2+ entry, causing Ca2+-handling abnormalities [13, 14]. Oxidative stress also exerts crucial effects on organ damage by triggering ferroptosis [15]. Nrf2 is a critical antioxidant transcription factor [16] and regulates the expression of multiple genes in the process of ferroptosis, such as ferritin heavy chain 1 (FTH1) [17], glutathione peroxide 4 (GPX4) [18, 19], and solute carrier family 7 member 11 (SLC7A11) [20, 21]. Therefore, oxidative stress may be a central event during DOX-induced myocardial injury by triggering apoptosis [22], ferroptosis [23], and pyroptosis [24].

Ferroptosis is a recently identified iron-driven cell death characterized by elevated lipid peroxidation, ROS overload, and plasma membrane rupture [8]. Several genes involved in ferroptosis have been identified, such as NADPH oxidase 1 (NOX1), cyclooxygenase-2 (COX-2), ACSL4, PTGS2, GPX4, FTH1, and SLC7A11 [25]. Ferroptosis is involved in DOX-induced cardiotoxicity [26]. Fang et al. demonstrated that ferrostatin-1 (Fer-1), a potent ferroptosis inhibitor, alleviates DOX-induced mortality, whereas pharmacological intervention of apoptosis, necroptosis, or autophagy could not significantly improve survival in DOX-treated mice [26].

CA, a bioactive component isolated from Cinnamonum cassia, possesses potent anti-oxidative [27, 28], anti-inflammatory [29], and anti-tumor capabilities [30]. Huang et al. demonstrated that CA increases phase II detoxifying enzyme expression by activating Nrf2 in hepatocellular carcinoma cells [27]. Kim et al. reported that CA alleviates dental pulp cell oxidative stress via activating the Nrf2/HO-1 pathway [28]. In the study, the biological effect of CA on alleviating DOX-induced cardiotoxicity was evaluated in vitro and in vivo.

Materials and methods

Cell culture and treatment

H9c2 cells were obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China) and cultivated in DMEM (GIBCO, NY, USA) containing 10% FBS (GIBCO) in a humidified CO2 incubator at 37 °C. H9c2 cells were seeded into 96-well plates (5000 cells per well) or 6-well plates (2 × 106 cells per well) overnight and then treated with DOX (0.5, 1, 2, 4, 8, and 10 μM) for 24 h.

Rat model of DOX-induced myocardial injury

The protocol was performed with the approval of the Experimental Animal Committee of Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine (No. PZSHUTCM210312009) according to ARRIVE guidelines [31] to decrease animals suffering. Male Sprague-Dawley rats (approximately 8 weeks old) were obtained from the Shanghai Model Organisms Center, Inc. (China) and maintained in specific pathogen-free facilities (temperature: 21–24°C) with water and food ad libitum. In the DOX group, rats (n = 5) were injected intraperitoneally (i.p.) with DOX at doses of 15 mg/kg (DOX was dissolved in 0.9% saline) as previously described [32]. In the CA treatment group, after treatment with 15 mg/kg of DOX, rats (n = 5) were additionally gavaged with CA at a dose of 50 mg/kg for 6 weeks, as previously described [33, 34] and in our preliminary experiments. In the control group (n = 5), rats were injected i.p. with 0.9% saline. Subsequently, rats were euthanized by inhalation of 2% isoflurane to collect blood and heart tissues for subsequent analysis.

Cell viability

The CCK-8 reagent (Abcam, CA, USA) was applied to measure H9c2 cell viability. Briefly, H9c2 cells were cultured in 96-well plates (5×103 cells per well) and treated with DOX (0.5, 1, 2, 4, 8, and 10 μM) for 24 h in the presence or absence of CA (0, 20, 40, 80, 100, 160, and 200 μM), Fer-1 (1 μM), or Erastin (10 μM). After that, cells were incubated with CCK-8 (10 μL) for 60 min in a humidified CO2 incubator, and then absorbance at 450 nm was read with a DR-200Bc microplate reader (BIOBASE, Shangdong, China).

TUNEL assay

TUNEL was used to assess H9c2 cell death. In brief, cells were seeded into 12-well plates and then treated with 4 μM of DOX and 100 μM of CA. After treatment for 24 h, cells were fixed with 4% PFA and stained with TUNEL (Beyotime, Shanghai, China) for 60 min. The fluorescence signal was captured with a XSPY-3201LED fluorescent microscope (CSOIF, Shanghai, China).

Serum biochemical indexes

Several serum biochemical indexes, including CK and LDH, were assessed using commercial kits (CK: ab155901, Abcam, CA, USA; LDH: ab102526, Abcam) in line with the manufacturer’s specifications.

Intracellular ROS assay

H9c2 cells cultured in 12-well plates (0.1×105 cells / well) were treated with 4 μM of DOX and 100 μM of CA for 24 h. After washing thrice in PBS, cells were incubated with 10 μM of DCFH-DA (MedChem Express, NJ, USA) for 20 min in the dark. The fluorescence signal was captured with a XSPY-3201LED fluorescent microscope (CSOIF, Shanghai, China).

Assessment of MDA, SOD, GSH, and GSH-Px

Cardiac samples were homogenized in RIPA buffer on ice, and then the supernatant was collected from tissue lysates via centrifuging at 3000 rpm for 15 min at 4°C. MDA, SOD, GSH, and GSH-Px levels in the supernatant were assessed using commercial kits in line with the manufacturer’s specifications.

Hematoxylin-eosin (HE) and Masson staining

Cardiac tissues were fixed with 4% PFA, embedded in paraffin, serially sectioned, and stained with HE and Masson (Beyotime). Images were examined using an N-800M light microscope (OLABO, Shandong, China).

Quantitative real-time PCR (qRT-PCR)

Total RNA was collected with TRIzol (Beyotime), and first-strand cDNA was synthesized with M-MLV reverse transcriptase (Sigma-Aldrich, MO, USA) and Oligo (dT)18 primers. qRT-PCR was performed with BeyoFast SYBR Green qPCR Mix (Beyotime) on a StepOnePlus real-time PCR system (Thermo Fisher Scientific). The temperature procedure is: 10 min at 95°C followed by 35 cycles of 95°C for 25 s and 58°C for 10 s. β-actin was applied as a reference gene. The 2(-ΔΔCT) method was applied to calculate the relative mRNA level [35]. All primers were shown in S1 Table.

Western blot

Total protein was collected from tissues or cells with RIPA buffer (Thermo Fisher Scientific). Nuclear and cytoplasmic proteins were isolated from cardiac tissues or H9c2 cells using a NE-PER reagent (Thermo Fisher Scientific) according to the manufacturer’s protocol. Equal amounts of total protein (approximately 30 μg) were separated using 10% SDS-PAGE and transferred to PVDF membranes (Merck, MA, USA). After blocking with 5% no-fat milk, membranes were incubated with antibodies against Nrf2 (1:1000, ab92946, Abcam), HO-1 (1:4000, ab68477), Gpx4 (1:3500, ab125066), Acsl4 (1:10000, ab155282), Ptgs2 (1:3000, ab179800), β-actin (1:4000, ab8226), and Lamin B1 (1:800, ab229025) overnight at 4°C. Then membranes were incubated with HRP-labeled secondary antibodies after washing three times in TBST. Immunoblot was observed using an enhanced chemiluminescence assay (Beyotime).

Statistics

The data were shown as the mean ± SD from three separate experiments. GraphPad Prism 7.0 (CA, USA) was used to compare the difference between two groups using the student’s t-test or among multiple groups using a one-way ANOVA followed by the Scheffé test. The difference was statistically significant when p < 0.05.

Results

CA alleviated DOX-induced myocardial injury

Consistent with previous studies [11, 36], DOX exhibited a dose-dependent cytotoxic effect on H9c2 cells (Fig 1A). Based on the results, 4 μM of DOX was applied to treat H9c2 cells in subsequent experiments. The cytotoxic effects of CA on H9c2 cells were also assessed using the CCK-8 assay. Fig 1B showed that CA did not exhibit a significant cytotoxic effect on H9c2 cells up to 100 μM. Therefore, 100 μM of CA was applied to treat H9c2 cells in subsequent experiments. The protective role of CA against DOX-induced cardiotoxicity was next assessed. As shown in Fig 1C, CA significantly reversed the effect of DOX on inhibiting H9c2 cell viability. CA also alleviated DOX-induced H9c2 cell death (Fig 1D and 1E). Furthermore, DOX treatment increased serum CK and LDH levels in rats, whereas these effects were reversed by CA (Fig 1F and 1G). The results from HE staining showed that DOX resulted in an obvious cardiomyocyte injury, as suggested by misaligned muscle fibers and widened intercellular spaces, whereas these effects were blocked by CA (Fig 1H). Masson staining revealed that DOX increased collagen fibers in the myocardial interstitium, and their distribution was chaotic, whereas myocardial fibrosis was obviously improved and tended to normalize after CA treatment (Fig 1I). CA also decreased DOX-induced Col1α1 and α-SMA expression in heart tissues (Fig 1J). These results demonstrate that DOX exhibits serious cardiotoxicity in vitro and in vivo, which can be reversed by CA.

thumbnail
Fig 1. CA alleviated DOX-induced myocardial injury in vitro and in vivo.

H9c2 cells were treated with different doses of DOX (A) or CA (B), and then cell viability was measured using CCK8 after 24 h (n = 3). (C) H9c2 cells were treated with 4 μM of DOX and 100 μM of CA for 24 h, and cell viability was measured using CCK8 after 24 h (n = 3). (D and E) H9c2 cells were treated with 4 μM of DOX and 100 μM of CA for 24 h, and cell death was measured using TUNEL (n = 3). Rats were treated with 15 mg/kg of DOX with or without CA (50 mg/kg, 6 weeks), and serum CK (F) and LDH (G) levels were assessed using commercial kits (n = 5). (H) HE staining was used to assess myocardial injury in rats treated with DOX and CA (n = 5). (I) Masson staining was used to assess myocardial fibrosis in rats treated with DOX and CA (n = 5). qRT-PCR was performed to assess Col1α1 and α-SMA (J) mRNA levels in rats treated with DOX and CA (n = 5). *p<0.05, **p<0.01.

https://doi.org/10.1371/journal.pone.0292124.g001

CA alleviated DOX-induced myocardial oxidative stress

Given that oxidative stress is a major mechanism of DOX-induced myocardial injury, we next investigated whether CA alleviated DOX-induced cardiotoxicity by decreasing oxidative stress. Fig 2A and 2B revealed that DOX obviously increased ROS levels in H9c2 cells, whereas CA repressed DOX-induced ROS. In a rat model of DOX-induced myocardial injury, DOX treatment suppressed SOD, GSH, and GSH-Px levels and increased MDA levels in cardiac tissues, whereas the effect was blocked by CA (Fig 2C–2F). These data suggest that CA effectively represses DOX-triggered myocardial oxidative stress in vitro and in vivo.

thumbnail
Fig 2. CA alleviated DOX-induced myocardial oxidative stress.

(A and B) H9c2 cells were incubated with DOX (4 μM) and CA (100 μM), and intracellular ROS levels were measured using the DCFH-DA assay after 24 h (n = 3). MDA (C), SOD (D), GSH (E), and GSH-px (F) levels in cardiac tissues were assessed using commercial kits (n = 5). *p<0.05, **p<0.01.

https://doi.org/10.1371/journal.pone.0292124.g002

CA activated Nrf2/HO-1 signaling

Nrf2 exerts a critical role in the anti-oxidative defense by increasing anti-oxidant enzyme expression [37, 38], and CA possesses anti-oxidative capacity by promoting nuclear translocation of Nrf2 in human renal mesangial cells and dental pulp cells [28, 39]. Therefore, we further investigated whether CA alleviated myocardial oxidative stress by activating Nrf2/HO-1 signaling. Fig 3A–3C revealed that DOX markedly repressed nuclear translocation of Nrf2, as suggested by increased cytoplasmic Nrf2 levels and decreased nuclear Nrf2 levels. More importantly, Nrf2 was reactivated and tended to normalize after CA treatment (Fig 3A–3C). Then we investigated whether CA-induced Nrf2 activation further facilitated HO-1 expression, an Nrf2 target gene. Fig 3D–3F showed that DOX treatment suppressed HO-1 expression at the mRNA and protein levels in H9c2 cells, whereas the effect was blocked by CA in a time-dependent manner. The regulatory roles of CA in Nrf2/HO-1 signaling were further validated in animal models. Although CA did not restore the total Nrf2 levels in DOX-treated rats (Fig 4A and 4B), CA accelerated the nuclear translocation of Nrf2 (Fig 4C and 4D). Moreover, HO-1 expression was restored by CA in DOX-treated rats (Fig 4A and 4B).

thumbnail
Fig 3. CA activated Nrf2/HO-1 signaling in vitro.

Western blot assay (A) and quantification (C) of cytoplasmic Nrf2 protein levels in H9c2 cells treated with DOX (4 μM) and CA (100 μM) (n = 3). Western blot assay (B) and quantification (C) of nuclear Nrf2 protein levels in H9c2 cells treated with DOX (4 μM) and CA (100 μM) (n = 3). (D) qRT-PCR assay of HO-1 mRNA levels in H9c2 cells treated with DOX (4 μM) and CA (100 μM) (n = 3). Western blot assay (E) and quantification (F) of cytoplasmic HO-1 protein levels in H9c2 cells treated with DOX (4 μM) and CA (100 μM) (n = 3). **p<0.01.

https://doi.org/10.1371/journal.pone.0292124.g003

thumbnail
Fig 4. CA activated Nrf2/HO-1 signaling in vivo.

(A and B) The total protein levels of Nrf2 and HO-1 in different groups of cardiac tissues (n = 5) were assessed using western blot assay. (C and D) The nuclear protein levels of Nrf2 in different groups of cardiac tissues (n = 5) were assessed using western blot assay.

https://doi.org/10.1371/journal.pone.0292124.g004

CA increased cardiomyocyte viability by repressing ferroptosis

Nrf2 signaling is closely correlated with ferroptosis [40], which exerts an important role in myocardial oxidative injury [41, 42]. To assess the role of DOX and CA in ferroptosis, H9c2 cells were treated with DOX and CA, and then ferroptosis markers (iron concentration, ROS, MDA, and GSH levels) were examined. Fig 5A revealed that iron concentration was significantly increased in H9c2 cells after DOX treatment, whereas the effect was blocked by CA. The role of CA in repressing DOX-induced ferroptosis was also verified by assessing ROS, MDA, and GSH levels (Fig 2A, 2B and 2D). Furthermore, ferroptosis-related gene (Gpx4, Acsl4, and Ptgs2) levels were assessed in H9c2 cells after treatment with DOX and CA. Fig 5B showed DOX down-regulated Gpx4 mRNA levels and up-regulated Ptgs2 and Acsl4 mRNA levels, whereas CA reversed these effects. Western blot assay further revealed that DOX decreased Gpx4 protein expression and increased Ptgs2 and Acsl4 protein expression, whereas these effects were blocked by CA (Fig 5C–5F), indicating that CA repressed DOX-induced ferroptosis. Functionally, Fer-1 increased DOX-treated H9c2 cell viability like CA (Fig 5G). More importantly, Erastin, a selective ferroptosis activator, destroyed the effect of CA on restoring H9c2 cell viability (Fig 5G). These results demonstrate that ferroptosis is correlated with DOX-induced cardiotoxicity, and CA alleviates DOX-induced cardiotoxicity by inhibiting Nrf2-dependent ferroptosis.

thumbnail
Fig 5. CA increased cardiomyocyte viability through repressing ferroptosis.

(A) Relative iron concentration was assessed using commercial kits in H9c2 cells treated with DOX (4 μM) and CA (100 μM) (n = 3). (B) qRT-PCR assay of Gpx4, Ptgs2, and Acsl4 mRNA levels in H9c2 cells treated with DOX (4 μM) and CA (100 μM) (n = 3). Western blot assay (C) and quantification of Gpx4 (D), Acsl4 (E), and Ptgs2 (F) protein levels in H9c2 cells treated with DOX (4 μM) and CA (100 μM) (n = 3). (G) H9c2 cells were treated with 4 μM of DOX in the presence or absence of CA (100 μM), Fer-1 (1 μM), or Erastin (10 μM) for 24 h, and cell viability was measured with CCK8 (n = 3). *p<0.05, **p<0.01.

https://doi.org/10.1371/journal.pone.0292124.g005

Discussion

Despite its efficacy against many malignancies, DOX exhibits severe cardiotoxicity, affecting approximately 30% of patients within five years after treatment [43]. Given that heart failure is a dominant reason for non-cancerous death after DOX treatment [43], relieving or eliminating DOX-triggered myocardial injury is essential to increasing its clinical application. Herein, we revealed the effect of CA on relieving DOX-induced cardiotoxicity. The main findings of this study were that: i) CA attenuated DOX-induced myocardial injury; ii) CA attenuated DOX-induced myocardial oxidative stress; iii) CA re-activated Nrf2/HO-1 signaling; and iv) CA enhanced cardiomyocyte viability by repressing cardiomyocyte ferroptosis.

Hydroxyl radicals, produced in physiological and pathological situations, are detoxified by GSH, which is a necessary antioxidant to maintain redox homeostasis through recycling enzymatic (GPXs, SOD, GST) and non-enzymatic (vitamin E) antioxidants [44]. DOX frequently results in GSH depletion through dysregulation of nicotinamide adenine dinucleotide phosphate (NADPH) [44]. NADPH is applied as the substrate of NADPH oxidases (NOXs) to generate ROS following DOX treatment [45]. Given that DOX-induced ROS overgeneration is a vital pathogenic event in cardiomyocyte injury [46], the roles of CA in alleviating DOX-induced oxidative stress and subsequent ROS-triggered cardiomyocyte ferroptosis were investigated. In the study, we demonstrated that DOX treatment increased ROS levels in H9c2 cells, whereas CA repressed DOX-induced ROS. Furthermore, DOX decreased SOD, GSH, and GSH-Px levels in the heart tissues of DOX-treated rats and increased MDA levels, whereas the effect was blocked by CA, indicating that CA effectively represses DOX-triggered myocardial oxidative stress in vitro and in vivo. CA further mitigated DOX-triggered cardiomyocyte ferroptosis. These results suggest that CA is a promising agent for decreasing the side effects of DOX by inhibiting ferroptosis.

Mounting studies have revealed the biological role of plant-derived natural compounds in regulating ROS production. Emodin, an anthraquinone extracted from rhubarb, exhibits a protective role against myocardial infarction through suppressing ROS generation [47]. Allicin sensitizes hepatocellular carcinoma (HCC) cells to 5-FU by further increasing ROS levels in cancer cells [48]. As an antioxidant, allicin relieves trastuzumab-induced cardiotoxicity by decreasing ROS-mediated myocardial cell apoptosis [49]. Astragaloside IV, an active component in Astragalus membranaceus, attenuates adriamycin-induced myocardial fibrosis through repressing cardiac ferroptosis and ROS levels [50].

The above results suggest that plant-derived natural ingredients possess the potential to relieve DOX-triggered myocardial injury by inhibiting DOX-induced oxidative stress. Given the biological role of CA in increasing Nrf2 nuclear translocation [27, 28] and repressing oxidative stress [28], we explored whether CA can alleviate DOX-induced cardiotoxicity through decreasing myocardial oxidative stress. The current results demonstrated that CA relieved DOX-induced myocardial oxidative stress by assessing ROS, MDA, SOD, and GSH levels. Mechanistically, CA accelerated the nuclear translocation of Nrf2 and increased HO-1 expression in cardiomyocytes. Furthermore, we found that CA repressed DOX-induced cardiomyocyte ferroptosis, while Erastin treatment destroyed the effect of CA on increasing cardiomyocyte viability. The major limitations of this study were that, i) besides ferroptosis, other ROS-triggered cell death forms (apoptosis, pyroptosis, and necrocytosis) were regulated by CA, and ii) it is essential to further investigate how Nrf2 signaling was activated by CA.

Conclusions

These results suggest that CA alleviates DOX-induced cardiotoxicity, providing a promising opportunity to increase the clinical application of DOX.

Acknowledgments

We would like to thank all the authors who contributed papers to this collection. We would also like to thank the PLOS ONE staff for their valuable support.

References

  1. 1. Silber JH, Barber G. Doxorubicin-induced cardiotoxicity. The New England journal of medicine. 1995;333(20):1359–60. Epub 1995/11/16. pmid:7566043
  2. 2. Tokarska-Schlattner M, Zaugg M, Zuppinger C, Wallimann T, Schlattner U. New insights into doxorubicin-induced cardiotoxicity: the critical role of cellular energetics. Journal of molecular and cellular cardiology. 2006;41(3):389–405. Epub 2006/08/02. pmid:16879835
  3. 3. Caso S, Maric D, Arambasic M, Cotecchia S, Diviani D. AKAP-Lbc mediates protection against doxorubicin-induced cardiomyocyte toxicity. Biochimica et biophysica acta Molecular cell research. 2017;1864(12):2336–46. Epub 2017/09/20. pmid:28923249
  4. 4. Shinlapawittayatorn K, Chattipakorn SC, Chattipakorn N. The effects of doxorubicin on cardiac calcium homeostasis and contractile function. Journal of cardiology. 2022. Epub 2022/01/29. pmid:35086744
  5. 5. Zhang X, Hu C, Kong CY, Song P, Wu HM, Xu SC, et al. FNDC5 alleviates oxidative stress and cardiomyocyte apoptosis in doxorubicin-induced cardiotoxicity via activating AKT. Cell death and differentiation. 2020;27(2):540–55. Epub 2019/06/19. pmid:31209361
  6. 6. Ye X, Li Y, Lv B, Qiu B, Zhang S, Peng H, et al. Endogenous Hydrogen Sulfide Persulfidates Caspase-3 at Cysteine 163 to Inhibit Doxorubicin-Induced Cardiomyocyte Apoptosis. Oxidative medicine and cellular longevity. 2022;2022:6153772. Epub 2022/05/17. pmid:35571249
  7. 7. Wang Y, Yan S, Liu X, Deng F, Wang P, Yang L, et al. PRMT4 promotes ferroptosis to aggravate doxorubicin-induced cardiomyopathy via inhibition of the Nrf2/GPX4 pathway. Cell death and differentiation. 2022. Epub 2022/04/07. pmid:35383293
  8. 8. Kitakata H, Endo J, Ikura H, Moriyama H, Shirakawa K, Katsumata Y, et al. Therapeutic Targets for DOX-Induced Cardiomyopathy: Role of Apoptosis vs. Ferroptosis. International journal of molecular sciences. 2022;23(3). Epub 2022/02/16. pmid:35163335
  9. 9. Zhai J, Tao L, Zhang S, Gao H, Zhang Y, Sun J, et al. Calycosin ameliorates doxorubicin-induced cardiotoxicity by suppressing oxidative stress and inflammation via the sirtuin 1-NOD-like receptor protein 3 pathway. Phytotherapy research: PTR. 2020;34(3):649–59. Epub 2019/12/21. pmid:31858651
  10. 10. Zhang S, Liu X, Bawa-Khalfe T, Lu LS, Lyu YL, Liu LF, et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nature medicine. 2012;18(11):1639–42. Epub 2012/10/30. pmid:23104132
  11. 11. Zhao L, Qi Y, Xu L, Tao X, Han X, Yin L, et al. MicroRNA-140-5p aggravates doxorubicin-induced cardiotoxicity by promoting myocardial oxidative stress via targeting Nrf2 and Sirt2. Redox biology. 2018;15:284–96. Epub 2018/01/06. pmid:29304479
  12. 12. Kong CY, Guo Z, Song P, Zhang X, Yuan YP, Teng T, et al. Underlying the Mechanisms of Doxorubicin-Induced Acute Cardiotoxicity: Oxidative Stress and Cell Death. International journal of biological sciences. 2022;18(2):760–70. Epub 2022/01/11. pmid:35002523
  13. 13. Dhalla NS, Temsah RM, Netticadan T. Role of oxidative stress in cardiovascular diseases. Journal of hypertension. 2000;18(6):655–73. Epub 2000/06/29. pmid:10872549
  14. 14. Dhalla NS, Elimban V, Bartekova M, Adameova A. Involvement of Oxidative Stress in the Development of Subcellular Defects and Heart Disease. Biomedicines. 2022;10(2). Epub 2022/02/26. pmid:35203602
  15. 15. Tang D, Kroemer G. Ferroptosis. Current biology: CB. 2020;30(21):R1292–R7. Epub 2020/11/04.
  16. 16. Lu Z, Lu Q, Li Z. [Research progress of nuclear factor E 2-related factor 2 signaling pathway in neuroprotective mechanism of cerebral ischemia/reperfusion injury]. Zhonghua wei zhong bing ji jiu yi xue. 2022;34(3):325–8. Epub 2022/05/17. pmid:35574756
  17. 17. Sun X, Ou Z, Chen R, Niu X, Chen D, Kang R, et al. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 2016;63(1):173–84. Epub 2015/09/26. pmid:26403645
  18. 18. Dai C, Chen X, Li J, Comish P, Kang R, Tang D. Transcription factors in ferroptotic cell death. Cancer gene therapy. 2020;27(9):645–56. Epub 2020/03/04. pmid:32123318
  19. 19. Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox biology. 2019;23:101107. Epub 2019/01/30. pmid:30692038
  20. 20. Dong H, Qiang Z, Chai D, Peng J, Xia Y, Hu R, et al. Nrf2 inhibits ferroptosis and protects against acute lung injury due to intestinal ischemia reperfusion via regulating SLC7A11 and HO-1. Aging. 2020;12(13):12943–59. Epub 2020/07/01. pmid:32601262
  21. 21. Sasaki H, Sato H, Kuriyama-Matsumura K, Sato K, Maebara K, Wang H, et al. Electrophile response element-mediated induction of the cystine/glutamate exchange transporter gene expression. The Journal of biological chemistry. 2002;277(47):44765–71. Epub 2002/09/18. pmid:12235164
  22. 22. Liu D, Ma Z, Di S, Yang Y, Yang J, Xu L, et al. AMPK/PGC1alpha activation by melatonin attenuates acute doxorubicin cardiotoxicity via alleviating mitochondrial oxidative damage and apoptosis. Free radical biology & medicine. 2018;129:59–72. Epub 2018/09/03.
  23. 23. Sun L, Wang H, Xu D, Yu S, Zhang L, Li X. Lapatinib induces mitochondrial dysfunction to enhance oxidative stress and ferroptosis in doxorubicin-induced cardiomyocytes via inhibition of PI3K/AKT signaling pathway. Bioengineered. 2022;13(1):48–60. Epub 2021/12/14. pmid:34898356
  24. 24. Zhang L, Jiang YH, Fan C, Zhang Q, Li Y, Xue YT. MCC950 attenuates doxorubicin-induced myocardial injury in vivo and in vitro by inhibiting NLRP3-mediated pyroptosis. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. 2021;143:112133. Epub 2021/09/03. pmid:34474337
  25. 25. Distefano AM, Lopez GA, Bauer V, Zabaleta E, Pagnussat GC. Ferroptosis in plants: regulation of lipid peroxidation and redox status. The Biochemical journal. 2022;479(7):857–66. Epub 2022/04/20. pmid:35438135
  26. 26. Fang X, Wang H, Han D, Xie E, Yang X, Wei J, et al. Ferroptosis as a target for protection against cardiomyopathy. Proceedings of the National Academy of Sciences of the United States of America. 2019;116(7):2672–80. Epub 2019/01/30. pmid:30692261
  27. 27. Huang TC, Chung YL, Wu ML, Chuang SM. Cinnamaldehyde enhances Nrf2 nuclear translocation to upregulate phase II detoxifying enzyme expression in HepG2 cells. Journal of agricultural and food chemistry. 2011;59(9):5164–71. Epub 2011/04/08. pmid:21469739
  28. 28. Kim NY, Ahn SG, Kim SA. Cinnamaldehyde protects human dental pulp cells against oxidative stress through the Nrf2/HO-1-dependent antioxidant response. European journal of pharmacology. 2017;815:73–9. Epub 2017/09/09. pmid:28882558
  29. 29. Qi L, Mao H, Lu X, Shi T, Wang J. Cinnamaldehyde Promotes the Intestinal Barrier Functions and Reshapes Gut Microbiome in Early Weaned Rats. Frontiers in nutrition. 2021;8:748503. Epub 2021/10/30. pmid:34712688
  30. 30. Lee CW, Lee SH, Lee JW, Ban JO, Lee SY, Yoo HS, et al. 2-hydroxycinnamaldehyde inhibits SW620 colon cancer cell growth through AP-1 inactivation. Journal of pharmacological sciences. 2007;104(1):19–28. Epub 2007/05/19. pmid:17510524
  31. 31. Mashima T, Iwasaki R, Kawata N, Kawakami R, Kumagai K, Migita T, et al. In silico chemical screening identifies epidermal growth factor receptor as a therapeutic target of drug-tolerant CD44v9-positive gastric cancer cells. British journal of cancer. 2019;121(10):846–56. Epub 2019/10/15. pmid:31607750
  32. 32. Yuan YP, Ma ZG, Zhang X, Xu SC, Zeng XF, Yang Z, et al. CTRP3 protected against doxorubicin-induced cardiac dysfunction, inflammation and cell death via activation of Sirt1. Journal of molecular and cellular cardiology. 2018;114:38–47. Epub 2017/10/25. pmid:29061338
  33. 33. Qian D, Tian J, Wang S, Shan X, Zhao P, Chen H, et al. Trans-cinnamaldehyde protects against phenylephrine-induced cardiomyocyte hypertrophy through the CaMKII/ERK pathway. BMC complementary medicine and therapies. 2022;22(1):115. Epub 2022/04/27. pmid:35468773
  34. 34. Xiao Q. Cinnamaldehyde attenuates kidney senescence and injury through PI3K/Akt pathway-mediated autophagy via downregulating miR-155. Renal failure. 2022;44(1):601–14. Epub 2022/04/02. pmid:35361048
  35. 35. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–8. Epub 2002/02/16. pmid:11846609
  36. 36. Pereira GC, Silva AM, Diogo CV, Carvalho FS, Monteiro P, Oliveira PJ. Drug-induced cardiac mitochondrial toxicity and protection: from doxorubicin to carvedilol. Current pharmaceutical design. 2011;17(20):2113–29. Epub 2011/07/02. pmid:21718248
  37. 37. 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 biology. 2018;16:189–98. Epub 2018/03/11. pmid:29524841
  38. 38. Smith RE, Tran K, Smith CC, McDonald M, Shejwalkar P, Hara K. The Role of the Nrf2/ARE Antioxidant System in Preventing Cardiovascular Diseases. Diseases. 2016;4(4). Epub 2017/09/22. pmid:28933413
  39. 39. Zheng H, Whitman SA, Wu W, Wondrak GT, Wong PK, Fang D, et al. Therapeutic potential of Nrf2 activators in streptozotocin-induced diabetic nephropathy. Diabetes. 2011;60(11):3055–66. Epub 2011/10/26. pmid:22025779
  40. 40. Guo N, Chen Y, Zhang Y, Deng Y, Zeng F, Li X. Potential Role of APEX1 During Ferroptosis. Frontiers in oncology. 2022;12:798304. Epub 2022/03/22. pmid:35311089
  41. 41. Shen Y, Wang X, Shen X, Wang Y, Wang S, Zhang Y, et al. Geniposide Possesses the Protective Effect on Myocardial Injury by Inhibiting Oxidative Stress and Ferroptosis via Activation of the Grsf1/GPx4 Axis. Frontiers in pharmacology. 2022;13:879870. Epub 2022/05/24. pmid:35600863
  42. 42. Gao Z, Chen Z, Xiong Z, Liu X. Ferroptosis—A new target of osteoporosis. Experimental gerontology. 2022:111836. Epub 2022/05/23. pmid:35598699
  43. 43. Schirone L, D’Ambrosio L, Forte M, Genovese R, Schiavon S, Spinosa G, et al. Mitochondria and Doxorubicin-Induced Cardiomyopathy: A Complex Interplay. Cells. 2022;11(13). Epub 2022/07/10. pmid:35805084
  44. 44. Sangweni NF, Gabuza K, Huisamen B, Mabasa L, van Vuuren D, Johnson R. Molecular insights into the pathophysiology of doxorubicin-induced cardiotoxicity: a graphical representation. Archives of toxicology. 2022;96(6):1541–50. Epub 2022/03/26. pmid:35333943
  45. 45. Xiao W, Wang RS, Handy DE, Loscalzo J. NAD(H) and NADP(H) Redox Couples and Cellular Energy Metabolism. Antioxidants & redox signaling. 2018;28(3):251–72. Epub 2017/06/27. pmid:28648096
  46. 46. Zou L, Liang B, Gao Y, Ye T, Li M, Zhang Y, et al. Nicotinic Acid Riboside Regulates Nrf-2/P62-Related Oxidative Stress and Autophagy to Attenuate Doxorubicin-Induced Cardiomyocyte Injury. BioMed research international. 2022;2022:6293329. Epub 2022/03/05. pmid:35242876
  47. 47. Yu Y, Liu H, Yang D, He F, Yuan Y, Guo J, et al. Aloe-emodin attenuates myocardial infarction and apoptosis via up-regulating miR-133 expression. Pharmacological research. 2019;146:104315. Epub 2019/06/18. pmid:31207343
  48. 48. Liu H, Mao P, Wang J, Wang T, Xie CH. Allicin Protects PC12 Cells Against 6-OHDA-Induced Oxidative Stress and Mitochondrial Dysfunction via Regulating Mitochondrial Dynamics. Cellular physiology and biochemistry: international journal of experimental cellular physiology, biochemistry, and pharmacology. 2015;36(3):966–79. Epub 2015/06/20. pmid:26087780
  49. 49. Mousa AM, Soliman KEA, Alhumaydhi FA, Almatroudi A, Allemailem KS, Alsahli MA, et al. Could allicin alleviate trastuzumab-induced cardiotoxicity in a rat model through antioxidant, anti-inflammatory, and antihyperlipidemic properties? Life sciences. 2022;302:120656. Epub 2022/05/24. pmid:35605695
  50. 50. Luo LF, Guan P, Qin LY, Wang JX, Wang N, Ji ES. Astragaloside IV inhibits adriamycin-induced cardiac ferroptosis by enhancing Nrf2 signaling. Molecular and cellular biochemistry. 2021;476(7):2603–11. Epub 2021/03/04. pmid:33656642