Elsevier

Life Sciences

Volume 180, 1 July 2017, Pages 151-159
Life Sciences

(−)-Epigallocatechin-3-gallate alleviates doxorubicin-induced cardiotoxicity in sarcoma 180 tumor-bearing mice

https://doi.org/10.1016/j.lfs.2016.12.004Get rights and content

Abstract

Aims

(−)-Epigallocatechin-3-gallate (EGCG), a major green tea polyphenol compound, plays an important role in the prevention of cardiovascular disease and cancer. The present study aimed to investigate the effects of EGCG on doxorubicin (DOX)-induced cardiotoxicity in Sarcoma 180 (S180) tumor-bearing mice.

Main methods

S180 tumor-bearing mice were established by subcutaneous inoculation of S180 cells attached to the axillary region. The extent of myocardial injury was accessed by the amount of lactate dehydrogenase (LDH) released in serum. Heart tissue was morphologically studied with transmission electron microscopy. Apoptosis, reactive oxygen species (ROS) generation, mitochondrial membrane potential (ΔѰm) as well as calcium concentration were measured by flow cytometric analysis. Expression levels of manganese superoxide dismutase (MnSOD) were analyzed by Western blot.

Key findings

Results showed that the combination with EGCG and DOX significantly inhibited tumor growth and enhanced induction of apoptosis compared with DOX alone. Moreover, administration of EGCG could suppress DOX-induced cardiotoxicity as evidenced by alleviating LDH release and apoptosis in cardiomyocyte. EGCG-evoked cardioprotection was in association with the increase of ΔѰm and MnSOD expression. EGCG was also found to attenuate ROS generation and myocardial calcium overload in Sarcoma 180 tumor-bearing mice subjected to DOX.

Significance

EGCG alleviated DOX-induced cardiotoxicity possibly in part mediated by increasing of MnSOD and Ѱm, reducing myocardial calcium overload and subsequently attenuating the apoptosis and LDH release. Our findings suggest that co-administration of EGCG and DOX have potential as a feasible strategy to mitigate cardiotoxicity of DOX without compromising its chemotherapeutic value.

Introduction

Doxorubicin (DOX: also called adriamycin), anthracycline antibiotic, belongs to the broad spectrum and highly effective anticancer drug. It played a significant role in the treatment of several malignant tumors including leukemia, lymphoma and breast cancer. Although these beneficial anti-tumor effects of DOX are generally recognized, its severe adverse effects and complications still restrict the prognosis of cancer patients by the treatment of DOX [1], [2]. It is well established that the most common side effect is cardiotoxicity. It has been demonstrated that a high affinity between DOX and myocardial tissue is the main factor contributing to the DOX-induced severe cardiotoxicity in vivo. DOX-induced cardiotoxicity is in proportion to its administrative dose which often resulted in various cardiac arrhythmias and congestive heart failure. Currently, cumulative and irreversible cardiotoxicity is the major limiting factor of DOX in the clinical setting [3], [4], [5].

Oxidative stress resulting from quinone-generated reactive oxygen species (ROS) has been suggested to be a common mechanism involved in DOX-mediated cardiomtopathy. There contains abundant mitochondria in the myocardial tissue, accounting for approximately 40% of the total volume of the heart. Besides, DOX possesses a high affinity for mitochondrial cardiolipin, which resulted in the accumulation of DOX in cardiomyocytes [6], [7]. Under the conditions of DOX treatment, the heart becomes a location for redox reaction. Briefly, under the circumstances of NADPH, quinone structure of DOX is transformed into a seminquinone structure through one-electron reduction in the mitochondrial electron transport chain. Seminquinone structure of DOX could be returned to quinone structure via acting with O2 to generate a O2. The circulating of DOX between quinone structure and seminquinone structure was in association with the generation of O2, which further led to over-production of ROS. As a consequence, quinone-mediated over-production of ROS contributes to oxidative stress and cardiotoxicity under the conditions of DOX treatment [8], [9], [10].

Tea, the most regularly consumed beverage worldwide, received an extensive attention due to its health benefits. (−)-Epigallocatechin-3-gallate (EGCG), identified as a major green tea polyphenol, has been reported to exert a positive effect on cancer chemopreventive activities and cardiovascular support [11]. Chen et al. [12] found that the combination of DOX and EGCG enhanced the DOX-mediated anti-tumor effect in hepatocellular carcinoma cell lines in vitro. In addition, Saeed et al. [13] showed that EGCG preconditioning inhibited myocardial injury caused by DOX in rats, and the mechanism was closely related to its antioxidant activity. EGCG is generally well known for its antioxidant activity, cardioprotection, and anti-tumor effect. However, little is known about whether EGCG could exert protective effects against cardiomyopathy in the tumor-bearing mice associated by the anti-tumor treatment of DOX in vivo.

Owing to the health benefits of EGCG on the anticancer and cardioprotection activities, we hypothesised that it could be regarded as a synergistic agent in combination with DOX to improve the prognosis of cancer patients. Therefore, the present study was aimed to investigate the protective effects of EGCG against DOX-caused cardiotoxicity and further to illuminate the underlying mechanisms in Sarcoma 180 (S180) tumor-bearing mice.

Section snippets

Animals

Kunming mice [weighing 22.0 ± 2.0 g, Grade II, Certificate Number SCXK (gan) 2006-0001] were purchased from Jiangxi University of Traditional Chinese Medicine, Nanchang, China. Animals were acclimated to our laboratory environment for 1 week. They were housed under standard conditions (temperature 25 ± 2 °C, humidity 50 ± 10%,12 h light, 12 h dark), and were allowed free access to food and water. All animals used in this study were cared for in accordance with the Guidelines for the Care and Use of

Effects of EGCG on DOX-mediated anti-tumor effect in S180 tumor-bearing mice

As shown in Fig. 1A, S180 tumor-bearing mice were administrated with DOX alone or the mixture of DOX and EGCG, the tumor growth was found to be reduced. The combined regime was superior to DOX treatment alone. Additionally, we used flow cytometry analysis to quantify the apoptosis in murine S180 cells derived-tumor tissues. There was a significant increase of apoptosis in DOX group when compared with control group (Fig. 1B and C). As compared with the DOX group, the combination of EGCG + DOX

Discussion

Collateral injury, such as severe adverse effects and complications in conventional cancer chemotherapy generally caused the paradox referring to the cancer treatment by restricting therapeutic dosages and by impairing the outcome of patients. Cardiotoxicity is a representative complication among the DOX-induced collateral damages in noncancerous tissues [6], [18], [19]. Novel therapeutic strategies with new concepts are urgently desired to attenuate the cardiotoxicity of DOX without

Conclusion

In conclusion, it has been demonstrated for the first time that EGCG enhanced the anti-tumor activity of DOX and ameliorated DOX-induced myocardial oxidative stress in S180 tumor-bearing mice. EGCG-exerted heart benefits were related to the reduction of LDH release, apoptosis, ROS generation and calcium level, and the increase in △Ψm and expression of MnSOD (Fig. 8). Taken together, our findings supported that co-administration of EGCG and DOX could be considered as a feasible strategy to

Author contributions

“Ming-Yong Xie” contributed to conception and design of the study, generation and approval of the final version of the manuscript. “Yu-Fei Yao” contributed to generation, collection, assembly, analysis and/or interpretation of data, and drafting and revision of the manuscript. “Xiang Liu, Wen-Juan Li, Zi-Wei Shi, Yu-Xin Yan, Le-Feng Wang, and Ming Chen” contributed to generation, collection, assembly, analysis and/or interpretation of data.

Conflict of interest

All authors declare that there are no conflicts of interest.

Funding sources

The financial support for the study by the National Natural Science Foundation of China (31560460), Natural Science Foundation of Jiangxi Province (20132BAB214001 and 20151BAB204038), Natural Science Foundation of Education Department of Jiangxi Province (GJJ14217), and Youth Research Fund of State Key Laboratory of Food Science and Technology (SKLF-QN-201509) are gratefully acknowledged.

References (34)

  • O. Szenczi et al.

    Poly (ADP-ribose) polymerase regulates myocardial calcium handling in doxorubicin-induced heart failure

    Biochem. Pharmacol.

    (2005)
  • C. Gonçalves et al.

    Sensitizing osteosarcoma stem cells to doxorubicin-induced apoptosis through retention of doxorubicin and modulation of apoptotic-related proteins

    Life Sci.

    (2015)
  • S. Emamgholipour et al.

    Evidence for possible role of melatonin in reducing oxidative stress in multiple sclerosis through its effect on SIRT1 and antioxidant enzymes

    Life Sci.

    (2016)
  • S. Zhang et al.

    Identification of the molecular basis of doxorubicin-induced cardiotoxicity

    Nat. Med.

    (2012)
  • Y. Chen et al.

    Collateral damage in cancer chemotherapy: oxidative stress in nontargeted tissues

    Mol. Interv.

    (2007)
  • G. Liu et al.

    Spironolactone attenuates doxorubicin-induced cardiotoxicity in rats

    Cardiovasc. Ther.

    (2016)
  • R.C. Kukreja et al.

    The oxygen free radical system: from equations through membrane-protein interactions to cardiovascular injury and protection

    Cardiovasc. Res.

    (1992)
  • Cited by (38)

    • EGCG adjuvant chemotherapy: Current status and future perspectives

      2023, European Journal of Medicinal Chemistry
    • Epigallocatechin-3-gallate attenuates cyclophosphamide-induced damage in mouse ovarian tissue via suppressing inflammation, apoptosis, and expression of phosphorylated Akt, FOXO3a and rpS6

      2022, Reproductive Toxicology
      Citation Excerpt :

      Based on our results, 25 or 50 mg/kg EGCG had cytoprotective effects equal to those of N-acetylcysteine. Because EGCG enhances the anticancer activity of chemotherapeutic drugs on neoplastic cells without adverse effects against healthy cells [69–72], this catechin could be suitable for use as a therapeutic agent to efficiently prevent ovarian injury induced by chemotherapy. Lastly, we evaluated the immunoexpression of p-Akt, p-rpS6 and p-FOXO3a in follicles from mice treated with cyclophosphamide alone or combined with EGCG.

    View all citing articles on Scopus
    View full text