Hydrogen sulfide attenuates doxorubicin‑induced cardiotoxicity by inhibiting calreticulin expression in H9c2 cells

  • Authors:
    • Mi‑Hua Liu
    • Yuan Zhang
    • Xiao‑Long Lin
    • Jun He
    • Tian‑Ping Tan
    • Shao‑Jian Wu
    • Shan Yu
    • Li Chen
    • Yu‑Dan Chen
    • Hong‑Yun Fu
    • Cong Yuan
    • Jian Li
  • View Affiliations

  • Published online on: July 2, 2015     https://doi.org/10.3892/mmr.2015.4020
  • Pages: 5197-5202
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Doxorubicin (DOX) is a potent and currently available antitumor therapeutic agent; however, its clinical application is limited by the occurrence of cardiotoxicity. Preliminary evidence indicates that hydrogen sulfide (H2S) may exert protective effects against DOX cardiotoxicity. Therefore, the aim of the present study was to investigate whether calreticulin (CRT) is involved in the cardioprotection of H2S against DOX‑induced cardiotoxicity. DOX was observed to markedly induce injuries, including cytotoxicity and apoptosis, and also enhance the expression level of CRT. Notably, pretreatment of H9c2 cells with sodium hydrosulfide (a donor of H2S) significantly attenuated the decreased cell viability, the increased apoptosis rate and the increased expression level of CRT in H9c2 cells. In addition, pretreatment of H9c2 cells with N‑acetyl‑L‑cysteine, a scavenger of reactive oxygen species (ROS) prior to exposure to DOX, markedly decreased the expression of CRT. These results indicate that exogenous H2S attenuates DOX‑induced cardiotoxicity by inhibiting CRT expression in H9c2 cardiac cells.

Introduction

To date, doxorubicin (DOX) remains one of the most widely administered anticancer therapeutic agents, due to its potent therapeutic affects on a variety of cancer types, including leukemia, lymphoma and breast cancer (1). However, its clinical use is limited by severe toxic side-effects on the heart, which may lead to dilated cardiomyopathy and congestive heart failure (2). Numerous studies have shown that reactive oxygen species (ROS) generation has been implicated in DOX's cardiotoxicity, which ultimately leads to cardiomyocyte apoptosis (3). The signal transduction pathway that links DOX-induced oxidative stress and cardiac injuries is currently a topic of particular interest.

Calreticulin (CRT), a Ca2+-binding molecular chaperone in the endoplasmic reticulum (ER), is vital in cardiac physiology and pathology (4,5). Recently, CRT was identified as a novel embryonic cardiac gene, which is highly expressed in embryonic hearts (6), however, its expression is suppressed after birth (7). Postnatally, elevated levels of CRT expression lead to impaired development of the cardiac conductive system and may be responsible for complete heart block (8). In a study of transgenic mice overexpressing CRT in the heart, the mice developed bradycardia, associated with sinus node dysfunction, complete cardiac block and succumbed due to intractable heart failure (9). Furthermore, overexpression of CRT enhanced apoptosis in myocardial H9c2 cells under conditions of retinoic acid-induced differentiation (10) or oxidative stress (11). These findings indicate that CRT overexpression is a key factor determining cellular susceptibility to oxidative stress-induced apoptosis. Recent studies on H9c2 cells indicate that overexpression of CRT in cardiomyocytes affects the Akt signaling pathway and promotes apoptosis (10,12). However, the biological significance of CRT expression levels in DOX-induced cardiotoxicity currently remains unknown.

Hydrogen sulfide (H2S), a well-known toxic gas, has been specified as the third gasotransmitter along with nitric oxide and carbon monoxide (13). Accumulating evidence has shown that H2S exerts important physiologic and pathophysiological action in the regulation of cardiovascular function (14). Our previous study revealed that increased endogenous H2S generation in the early reperfusion phase is significant in ischemia preconditioning (IPC)-elicited protection in isolated hearts (14). Based on these previous studies, the present study investigates the role of CRT in the protective effects of H2S against DOX-induced cardiomyocyte injuries.

H9c2 cells were treated with 5 μM DOX to establish a chemotherapy-induced cardiotoxicity model (15) and the aim of the present study was to establish whether DOX induces expression of CRT in H9c2 cells. Furthermore, the role of CRT in the protective effect of H2S against DOX-induced injury in H9c2 cells was elucidated.

Materials and methods

Materials

Methyl thiazolyl-tetrazolium (MTT), Hoechst 33258, DOX, sodium hydrosulfide (NaHS), and N-acetyl-L-cysteine (NAC) were purchased from Sigma-Aldrich (St. Louis, MO, USA). All cell culture medium components were purchased from Thermo Fisher Scientific, Inc. (Waltham, MA, USA) unless otherwise stated. The H9c2 cardiac myocytes were obtained from the Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China) (originally from the American Type Culture Collection, Manassas, VA, USA).

Cell culture

H9c2 cardiac myocytes were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 μg/ml streptomycin and 100 U/ml penicillin streptomycin (both Gibco Life Technologies, Carlsbad, CA, USA) in a humidified 5% CO2 atmosphere at 37°C. The H9c2 cardiac myocytes were passaged every two days. H9c2 cardiac myocytes were seeded at a density of 2×106 cells/dish in 100-mm dishes with 10% fetal calf serum and incubated for 24 h, the culture medium was subsequently changed to 0.5% FBS DMEM for 24-h starvation.

MTT assay

The MTT assay is a standard method used to assess cell viability. Prior to each experiment, H9c2 cardiac myocytes (5,000 cells/well) were seeded in 96-well microtiter plates. Following incubation with NaHS for 30 min, the cells were treated with 5 μM DOX for a further 24 h. Subsequently, 10 μl MTT solution was added to each well and the plates were incubated for 4 h at 37°C in a 5% CO2. The absorbance was measured with the SpectraMax 190 Spectrophotometer (Molecular Devices LLC, Sunnyvale, CA, USA) at 470 nm and used to calculate the relative ratio of cell viability (optical density of treatment group/optical density of control group ×100%). Three independent experiments were performed for each experimental condition.

Assessment of cardiomyocyte cell apoptosis

Apoptosis was analyzed by fluorescence microscopy with the chromatin dye, Hoechst 33258. Following various treatments, the cells were fixed in ice-cold 4% paraformaldehyde dissolved in phosphate-buffered saline (PBS) at room temperature for 20 min. Non-specific binding was blocked using 5% normal goat serum in 0.01 M PBS containing 0.3% Triton X-100. Cells were washed twice with PBS and incubated with 10 μg/ml Hoechst 33258 for 15 min at room temperature in the dark. The cells were visualized under a fluorescence microscope (BX50-FLA; Olympus Corporation, Tokyo, Japan). Apoptotic cells exhibited condensed, fractured or distorted nuclei, whereas viable cells displayed normal nuclear size and uniform fluorescence.

Western blot analysis

Cells were homogenized directly into cell lysis buffer (Cell Signaling Technology, Inc., Danvers, MA, USA) and phosphatase inhibitor cocktail (Sigma-Aldrich), and lysates were centrifuged at 12,000 rpm for 10 min at 4°C. The protein concentration was determined with the use of a bicinchoninic acid protein assay kit according to the manufacturer's instruction. The extracted proteins were mixed with 5% sodium dodecyl sulfate (SDS)-PAGE sample buffer, then boiled at 100°C for 7 min and separated by electrophoresis on a 10% SDS-polyacrylamide gel. Subsequent to electrophoresis, proteins were transferred to polyvinylidene difluoride membranes. The membranes were blocked in Tris-buffered saline with 0.1% Tween-20 (TBS-T) containing 5% non-fat dry milk for 2 h at room temperature with rotation (20 rpm for 2 h). After blocking, the membranes were incubated with the following antibodies: Rabbit anti-cystathionine γ-lyase (CSE) polyclonal antibody (1:1,000; Cell Signaling Technology, Inc.) and rabbit CRT polyclonal antibody (1:200; Abcam, Cambridge, UK). Then, membranes were incubated in bovine serum albumin overnight at 4°C. The primary antibody was removed by washing the membranes three times in TBS-T and incubated for 2 h with the appropriate horseradish peroxidase-conjugated secondary antibodies. Following three washes in TBS-T, the antigen-antibody bands were detected using an Enhanced Chemiluminescence Reagent kit (Beyotime Institute of Biotechnology, Shanghai, China) and quantified using the Quantity One Software Package (Bio-Rad Laboratories, Ltd., Hemel Hempstead, UK).

Statistical analysis

Results are presented as the mean ± standard error of the mean. Statistical analysis was performed using Student's t-test or analysis of variance with SPSS 13.0 (SPSS Inc., Chicago, IL, USA) and P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of DOX on the expression of CRT in H9c2 cells

To investigate the effect of DOX on the expression of CRT, H9c2 cells were treated with 5 μM DOX for 0 (at the time of treatment), 3, 6, 12 and 24 h. Western blot analysis demonstrated that DOX treatment enhanced the CRT expression levels in a time-dependent manner (Fig. 1).

DOX inhibited the expression of CSE in H9c2 cells

CSE is the major enzyme responsible for endogenous H2S generation in H9c2 cells (16). Western blot analysis was performed to evaluate whether DOX decreases endogenous H2S production by inhibiting the expression of CSE. Treatment with 5 μM DOX for the indicated time periods (0, 3, 6, 12 and 24 h) caused a significant downregulation of CSE expression in H9c2 cells (Fig. 2). These data indicate that DOX induced the inhibition of CSE expression in H9c2 and contributed to the DOX-elicited decrease in endogenous H2S production.

Exogenous H2S inhibits DOX-induced expression of CRT in H9c2 cells

The effect of NaHS on the expressions of CRT induced by DOX was detected to assess whether the cyto-protective effect of H2S against DOX-induced toxicity was associated with the inhibition of CRT in H9c2 cells. The results demonstrated that pretreatment of H9c2 cells with 100 μM NaHS (a donor of H2S) for 30 min prior to exposure to 5 μmol/l DOX for 24 h significantly inhibited the DOX-induced overexpression of CRT (Fig. 3). These data indicate that the cardioprotection of H2S is associated with its inhibitory effect on DOX-induced CRT expression.

NAC suppresses the DOX-induced expression of CRT in H9c2 cells

To identify whether the inhibitory effect of NaHS on the DOX-induced increase in expression of CRT is associated with its antioxidation, H9c2 cells were pretreated with 1,000 μM NAC (an ROS scavenger) for 60 min prior to exposure to 5 μM DOX for 24 h. As shown in Fig. 4, similar to the inhibitory effect of NaHS pretreatment, the pretreatment of cells with NAC for 60 min markedly depressed the expression of CRT. The results revealed that the antioxidant effect, resulting from NAC administration, contributed to the inhibitory effect of H2S on the DOX-induced expression of CRT.

Effect of H2S and NAC on DOX-induced cytotoxicity

As presented in Fig. 5, exposure of H9c2 cells to DOX at a dose of 5 μM for 24 h induced marked cytotoxicity, leading to a decrease in cell viability. However, pretreatment of cells with 100 μM NaHS for 30 min prior to exposure to DOX significantly ameliorated the DOX-induced cytotoxicity, as evidenced by an increase in cell viability (P<0.01 compared with the DOX-treated group). Similar to the effect of NaHS, pretreatment with NAC for 60 min significantly attenuated the DOX-induced decrease in cell viability (P<0.01 compared with the DOX-treated group). Neither NaHS nor NAC alone altered cell viability in the H9c2 cells.

Effect of H2S and NAC on DOX-induced apoptosis

The effects of NaHS and NAC treatment on DOX-induced apoptosis was also observed. As shown in Fig. 6, H9c2 cells treated with 5 μM DOX for 24 h exhibited typical characteristics of apoptosis, including condensation of chromatin, shrinkage of nuclei and apoptotic bodies. However, pretreatment of cells with 100 μM NaHS for 30 min prior to DOX exposure markedly decreased the DOX-induced increase in cell number, as well as decreasing the nuclear condensation and fragmentation. In addition, H9c2 cells were preconditioned with a common ROS scavenger, NAC (1,000 μM) prior to DOX treatment. The results indicated that pretreatment of cells with NAC significantly attenuated DOX-induced H9c2 cell apoptosis. NaHS or NAC alone did not markedly alter the cell morphology or the percentage of apoptotic H9c2 cells. These findings suggest that an antioxidant effect participates in the inhibitory effect of H2S on the DOX-induced apoptosis of H9c2 cells.

Discussion

Numerous studies have revealed that the major molecular mechanism involved in DOX-induced cardiac toxicity is free radical-induced oxidative stress and cardiomyocyte death, as a result of apoptosis and necrosis. Consistent with previous studies (17,18), the present study observed that exposure of H9c2 cells to DOX markedly induced cellular injury, including decreased cell viability, as well as increased cell apoptosis and expression of CRT.

Previously, the cardioprotective effects of H2S have been demonstrated in animal models of disease (19). H2S infusion significantly reduces myocardial infract size and improves regional left ventricular function, as well as endothelium-dependent and -independent microvascular reactivity in a porcine model of myocardial ischemia-reperfusion (20). Furthermore, H2S attenuates myocardial necrosis and apoptosis (21). In addition, endogenous H2S is associated with the cardioprotection that results from metabolic inhibition preconditioning in rat ventricular myocytes (22). Inhibition of endogenous H2S generation, by its synthesis inhibitor (DL-propargylglycine), has been shown to block the protective effect of IPC in isolated hearts, as well as in isolated cardiac myocytes (23). In the present study, H9c2 cells were used to elucidate the effect of DOX on endogenous H2S generation. Exposure of H9c2 cells to DOX resulted in a significant decrease in H2S generation.

CRT is a major ER protein that is significant in cardiac development and pathology (24). Various reviews have revealed that CRT is highly expressed in embryonic hearts, but not in mature hearts, and may be an early cardiac gene product (25). Mice with a targeted disruption of the CRT gene succumb in utero exhibiting decreased ventricular cell mass due to increased apoptosis of cardiac myocytes (26). In addition, studies with CRT-deficient cells suggest that CRT participates in apoptosis (27). In the CRT transgenic heart, CRT-dependent cardiac block involves damage to L-type Ca2+ channels, and gap junction connexin-40 and -43, due to defective regulation of Ca2+ homeostasis (8). Overexpression of CRT suppresses Akt signaling and causes differentiation-induced apoptosis in H9c2 cells (10). In the present study, the results showed that the expression of CRT was increased following DOX treatment, and exogenous H2S preconditioning was demonstrated to suppress CRT expression while markedly attenuating DOX-induced apoptosis.

Increasing evidence indicates that ROS are significant in the pathogenesis of cardiac failure (28). Furthermore, antioxidants have been shown to exert protective and beneficial effects against heart failure (29). Oxidative stress is a primary mechanism by which DOX induces cardiomyocyte injury. Notably, the present study demonstrated that oxidative stress was involved in DOX-induced cell injury and established whether DOX activation of CRT is due to the induction of ROS. Pretreatment of H9c2 cells with NAC (a ROS scavenger) was shown to significantly attenuate DOX-induced expression of CRT. Thus, the results of the present study support the hypothesis that DOX induction of ROS activates CRT, which mediates DOX-induced injury in H9c2 cells.

In conclusion, the current study identified that H2S inhibits DOX-induced apoptosis in H9c2 cells, which may involve inhibition of ROS-mediated CRT expression. Therefore, the present study has elucidated the mechanisms of H2S-mediated anti-apoptosis in cardiomyocytes and provided evidence for identifying H2S as a candidate for application in the treatment of cardiovascular diseases.

Acknowledgments

The present study was supported by grants from the Medical Scientific Research Funds of Guangdong province (grant no. A2014810) and the Graduate Student Research Innovation Project of Hunan province (grant no. CX2013B397).

References

1 

Menna P, Recalcati S, Cairo G and Minotti G: An introduction to the metabolic determinants of anthracycline cardiotoxicity. Cardiovasc Toxicol. 7:80–85. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Lipshultz SE, Karnik R, Sambatakos P, Franco VI, Ross SW and Miller TL: Anthracycline-related cardiotoxicity in childhood cancer survivors. Curr Opin Cardiol. 29:103–112. 2014. View Article : Google Scholar

3 

Spallarossa P, Garibaldi S, Altieri P, Fabbi P, Manca V, Nasti S, Rossettin P, Ghigliotti G, Ballestrero A, Patrone F, et al: Carvedilol prevents doxorubicin-induced free radical release and apoptosis in cardiomyocytes in vitro. J Mol Cell Cardiol. 37:837–846. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Raturi A, Ortiz-Sandoval C and Simmen T: Redox dependence of endoplasmic reticulum (ER) Ca2+ signaling. Histol Histopathol. 29:543–552. 2014.

5 

Ma J and Pan Z: Retrograde activation of store-operated calcium channel. Cell Calcium. 33:375–384. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Mesaeli N, Nakamura K, Zvaritch E, Dickie P, Dziak E, Krause KH, Opas M, MacLennan DH and Michalak M: Calreticulin is essential for cardiac development. J Cell Biol. 144:857–868. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Milan D, Griffith J, Su M, Price ER and McKeon F: The latch region of calcineurin B is involved in both immunosuppressant-immunophilin complex docking and phosphatase activation. Cell. 79:437–447. 1994. View Article : Google Scholar : PubMed/NCBI

8 

Nakamura K, Robertson M, Liu G, Dickie P, Guo JQ, Duff HJ, Opas M, Kavanagh K and Michalak M: Complete heart block and sudden death in mice overexpressing calreticulin. J Clin Invest. 107:1245–1253. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Lynch JM, Chilibeck K, Qui Y and Michalak M: Assembling pieces of the cardiac puzzle; calreticulin and calcium-dependent pathways in cardiac development, health and disease. Trends Cardiovasc Med. 16:65–69. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Kageyama K, Ihara Y, Goto S, Urata Y, Toda G, Yano K and Kondo T: Overexpression of calreticulin modulates protein kinase B/Akt signaling to promote apoptosis during cardiac differentiation of cardiomyoblast H9c2 cells. J Biol Chem. 277:19255–19264. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Ihara Y, Urata Y, Goto S and Kondo T: Role of calreticulin in the sensitivity of myocardiac H9c2 cells to oxidative stress caused by hydrogen peroxide. Am J Physiol Cell Physiol. 290:C208–C221. 2006. View Article : Google Scholar

12 

Konishi M, Haraguchi G, Ohigashi H, Ishihara T, Saito K, Nakano Y and Isobe M: Adiponectin protects against doxorubicin-induced cardiomyopathy by anti-apoptotic effects through AMPK up-regulation. Cardiovasc Res. 89:309–319. 2011. View Article : Google Scholar

13 

Zhang Y, Tang ZH, Ren Z, Qu SL, Liu MH, Liu LS and Jiang ZS: Hydrogen sulfide, the next potent preventive and therapeutic agent in aging and age-associated diseases. Mol Cell Biol. 33:1104–1113. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Huang YE, Tang ZH, Xie W, Shen XT, Liu MH, Peng XP, Zhao ZZ, Nie DB, Liu LS and Jiang ZS: Endogenous hydrogen sulfide mediates the cardioprotection induced by ischemic post-conditioning in the early reperfusion phase. Exp Ther Med. 4:1117–1123. 2012.PubMed/NCBI

15 

Guo R, Lin J, Xu W, Shen N, Mo L, Zhang C and Feng J: Hydrogen sulfide attenuates doxorubicin-induced cardiotoxicity by inhibition of the p38 MAPK pathway in H9c2 cells. Int J Mol Med. 31:644–650. 2013.PubMed/NCBI

16 

Kimura H: Hydrogen sulfide: Its production, release and functions. Amino Acids. 41:113–121. 2011. View Article : Google Scholar

17 

Wang X, Wang XL, Chen HL, Wu D, Chen JX, Wang XX, Li RL, He JH, Mo L, Cen X, et al: Ghrelin inhibits doxorubicin cardiotoxicity by inhibiting excessive autophagy through AMPK and p38-MAPK. Biochem Pharmacol. 88:334–350. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Guo R, Wu K, Chen J, Mo L, Hua X, Zheng D, Chen P, Chen G, Xu W and Feng J: Exogenous hydrogen sulfide protects against doxorubicin-induced inflammation and cytotoxicity by inhibiting p38MAPK/NFκB pathway in H9c2 cardiac cells. Cell Physiol Biochem. 32:1668–1680. 2013.

19 

Ji Y, Pang QF, Xu G, Wang L, Wang JK and Zeng YM: Exogenous hydrogen sulfide postconditioning protects isolated rat hearts against ischemia-reperfusion injury. Eur J Pharmacol. 587:1–7. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Osipov RM, Robich MP, Feng J, Liu Y, Clements RT, Glazer HP, Sodha NR, Szabo C, Bianchi C and Sellke FW: Effect of hydrogen sulfide in a porcine model of myocardial ischemia-reperfusion: Comparison of different administration regimens and characterization of the cellular mechanisms of protection. J Cardiovasc Pharmacol. 54:287–297. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Sodha NR, Clements RT, Feng J, Liu Y, Bianchi C, Horvath EM, Szabo C and Sellke FW: The effects of therapeutic sulfide on myocardial apoptosis in response to ischemia-reperfusion injury. Eur J Cardiothorac Surg. 33:906–913. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Pan TT, Feng ZN, Lee SW, Moore PK and Bian JS: Endogenous hydrogen sulfide contributes to the cardioprotection by metabolic inhibition preconditioning in the rat ventricular myocytes. J Mol Cell Cardiol. 40:119–130. 2006. View Article : Google Scholar

23 

Bian JS, Yong QC, Pan TT, Feng ZN, Ali MY, Zhou S and Moore PK: Role of hydrogen sulfide in the cardioprotection caused by ischemic preconditioning in the rat heart and cardiac myocytes. J Pharmacol Exp Ther. 316:670–678. 2006. View Article : Google Scholar

24 

Michalak M, Lynch J, Groenendyk J, Guo L, Robert Parker JM and Opas M: Calreticulin in cardiac development and pathology. Biochim Biophys Acta. 1600:32–37. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Coe H and Michalak M: Calcium binding chaperones of the endoplasmic reticulum. Gen Physiol Biophys. 28:F96–F103. 2009.

26 

Rauch F, Prud'homme J, Arabian A, Dedhar S and St-Arnaud R: Heart, brain and body wall defects in mice lacking calreticulin. Exp Cell Res. 256:105–111. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Nakamura K, Bossy-Wetzel E, Burns K, Fadel MP, Lozyk M, Goping IS, Opas M, Bleackley RC, Green DR and Michalak M: Changes in endoplasmic reticulum luminal environment affect cell sensitivity to apoptosis. J Cell Biol. 150:731–740. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Schwarzer M, Osterholt M, Lunkenbein A, Schrepper A, Amorim P and Doenst T: Mitochondrial reactive oxygen species production and respiratory complex activity in rats with pressure overload-induced heart failure. J Physiol. 592:3767–3782. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Matsushima S, Ide T, Yamato M, Matsusaka H, Hattori F, Ikeuchi M, Kubota T, Sunagawa K, Hasegawa Y, Kurihara T, et al: Overexpression of mitochondrial peroxiredoxin-3 prevents left ventricular remodeling and failure after myocardial infarction in mice. Circulation. 113:1779–1786. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2015
Volume 12 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu MH, Zhang Y, Lin XL, He J, Tan TP, Wu SJ, Yu S, Chen L, Chen YD, Fu HY, Fu HY, et al: Hydrogen sulfide attenuates doxorubicin‑induced cardiotoxicity by inhibiting calreticulin expression in H9c2 cells. Mol Med Rep 12: 5197-5202, 2015
APA
Liu, M., Zhang, Y., Lin, X., He, J., Tan, T., Wu, S. ... Li, J. (2015). Hydrogen sulfide attenuates doxorubicin‑induced cardiotoxicity by inhibiting calreticulin expression in H9c2 cells. Molecular Medicine Reports, 12, 5197-5202. https://doi.org/10.3892/mmr.2015.4020
MLA
Liu, M., Zhang, Y., Lin, X., He, J., Tan, T., Wu, S., Yu, S., Chen, L., Chen, Y., Fu, H., Yuan, C., Li, J."Hydrogen sulfide attenuates doxorubicin‑induced cardiotoxicity by inhibiting calreticulin expression in H9c2 cells". Molecular Medicine Reports 12.4 (2015): 5197-5202.
Chicago
Liu, M., Zhang, Y., Lin, X., He, J., Tan, T., Wu, S., Yu, S., Chen, L., Chen, Y., Fu, H., Yuan, C., Li, J."Hydrogen sulfide attenuates doxorubicin‑induced cardiotoxicity by inhibiting calreticulin expression in H9c2 cells". Molecular Medicine Reports 12, no. 4 (2015): 5197-5202. https://doi.org/10.3892/mmr.2015.4020