Lab Anim Res.  2017 Jun;33(2):165-170. 10.5625/lar.2017.33.2.165.

Comparision of doxorubicin-induced cardiotoxicity in the ICR mice of different sources

Affiliations
  • 1College of Pharmacy, Pusan National University, Busan, Korea. youngjung@pusan.ac.kr
  • 2Department of Biomedical Laboratory Science, Daekyeung College, Gyeongsan, Korea.
  • 3College of Pharmacy, Kyungsung University, Busan, Korea.
  • 4Department of Microbiology and Immunology, INJE University College of Medicine, Busan, Korea.
  • 5Exercise Biochemistry Laboratory, Korea National Sport University, Seoul, Korea.
  • 6Department of Biomaterials Science, College of Natural Resources & Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea.
  • 7College of Veterinary Medicine, Kyungpook National University, Daegu, Korea.

Abstract

Doxorubicin is a widely used chemotherapeutic agents and is now part of standard therapeutic regimens for a variety of cancers (eg, hematopoietic malignancies and advanced solid tumors of the breast, ovary, thyroid, and bone). However, a potentially lethal and dose-dependent cardiotoxicity that appears within a short time after treatment limits the usage of doxorubicin in cancer patients. Although the mechanism of doxorubicin-induced cardiotoxicity is not completely understood, it is thought that free radical-induced oxidative stress and excessive production of reactive oxygen species are primary drivers of its toxicity. In this study, we compared the doxorubicin-induced cardiotoxicity of ICR mice obtained from three different sources and evaluated the utility of Korl:ICR stock established by the Korean FDA. Because doxorubicin-induced cardiotoxicity is thought to involve the excessive generation of ROS followed by oxidative stress, we determined the representative tissue index of oxidation, lipid peroxidation, and antioxidant, glutathione (GSH), as well as the parameters of heart injury. Doxorubicin treatment successfully induced cardiotoxicity as evidenced by histological examination and serum parameters (eg, levels of LDH and CK activities) in ICR mice. It was accompanied by increased lipid peroxidation and a decrease in both cysteine and GSH, further supporting previous reports that oxidative stress is a potential mechanism of doxorubicin-induced cardiotoxicity. Of interest, we did not observe a significant difference in doxorubicin-induced cardiotoxicity among mice of different origins. Collectively, our results suggest that Korl:ICR strain may be useful in the research of doxorubicin-induced cardiotoxicity.

Keyword

Cardiotoxicity; doxorubicin; oxidative stress; ICR mouse

MeSH Terms

Animals
Breast
Cardiotoxicity*
Cysteine
Doxorubicin
Female
Glutathione
Heart Injuries
Hematologic Neoplasms
Humans
Lipid Peroxidation
Mice
Mice, Inbred ICR*
Ovary
Oxidative Stress
Reactive Oxygen Species
Thyroid Gland
Cysteine
Doxorubicin
Glutathione
Reactive Oxygen Species

Figure

  • Figure 1 Effects of doxorubicin (DOX) treatment on LDH (A) and CK-MB (B) activities in the heart of ICR mice. **, *** Significantly different from the corresponding control mice (ANOVA followed by Newman-Keuls multiple range test, P<0.01, 0.001, respectively).

  • Figure 2 Histopathological changes in cardiac tissue following doxorubicin (DOX) treatment in ICR mice from three different sources. Pictures of heart sections from the control (A, C, and E) and DOX-treated (B, D, and F). Original magnification, ×100.

  • Figure 3 Effects of doxorubicin (DOX) treatment on MDA levels in the heart of ICR mice. *** Significantly different from the corresponding control mice (ANOVA followed by Newman-Keuls multiple range test, P<0.001).

  • Figure 4 Changes in the levels of cysteine (A) and GSH (B) in the heart of doxorubicin (DOX)-treated ICR mice. **, *** Significantly different from the corresponding control mice (ANOVA followed by Newman-Keuls multiple range test, P<0.01, 0.001, respectively).


Reference

1. Thavendiranathan P, Poulin F, Lim KD, Plana JC, Woo A, Marwick TH. Use of myocardial strain imaging by echocardiography for the early detection of cardiotoxicity in patients during and after cancer chemotherapy: a systematic review. J Am Coll Cardiol. 2014; 63(25 Pt A):2751–2768. PMID: 24703918.
2. Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev. 2004; 56(2):185–229. PMID: 15169927.
Article
3. Perrino C, Schiattarella GG, Magliulo F, Ilardi F, Carotenuto G, Gargiulo G, Serino F, Ferrone M, Scudiero F, Carbone A, Trimarco B, Esposito G. Cardiac side effects of chemotherapy: state of art and strategies for a correct management. Curr Vasc Pharmacol. 2014; 12(1):106–116. PMID: 22563720.
Article
4. Raschi E, Vasina V, Ursino MG, Boriani G, Martoni A, De Ponti F. Anticancer drugs and cardiotoxicity: Insights and perspectives in the era of targeted therapy. Pharmacol Ther. 2010; 125(2):196–218. PMID: 19874849.
Article
5. Carter SK. Adriamycin-Review. J Natl Cancer Inst. 1975; 55(6):1265–1274. PMID: 1107570.
6. Bristow MR, Mason JW, Billingham ME, Daniels JR. Doxorubicin Cardiomyopathy-Evaluation by Phonocardiography, Endomyocardial Biopsy, and Cardiac-Catheterization. Ann Intern Med. 1978; 88(2):168–175. PMID: 626445.
7. Bristow MR, Thompson PD, Martin RP, Mason JW, Billingham ME, Harrison DC. Early Anthracycline Cardiotoxicity. Am J Med. 1978; 65(5):823–832. PMID: 707541.
Article
8. Maksimenko AV, Vavaev AV. Antioxidant enzymes as potential targets in cardioprotection and treatment of cardiovascular diseases. Enzyme antioxidants: the next stage of pharmacological counterwork to the oxidative stress. Heart Int. 2012; 7(1):e3. PMID: 22690296.
Article
9. Todorova VK, Beggs ML, Delongchamp RR, Dhakal I, Makhoul I, Wei JY, Klimberg VS. Transcriptome profiling of peripheral blood cells identifies potential biomarkers for doxorubicin cardiotoxicity in a rat model. PLoS One. 2012; 7(11):e48398. PMID: 23209553.
Article
10. Sterba M, Popelova O, Vavrova A, Jirkovsky E, Kovarikova P, Gersl V, Simunek T. Oxidative stress, redox signaling, and metal chelation in anthracycline cardiotoxicity and pharmacological cardioprotection. Antioxid Redox Signal. 2013; 18(8):899–929. PMID: 22794198.
11. Thayer WS. Adriamycin Stimulated Superoxide Formation in Sub-Mitochondrial Particles. Chem Biol Interact. 1977; 19(3):265–278. PMID: 202411.
Article
12. Doroshow JH, Reeves J. Anthracycline-Enhanced Oxygen Radical Formation in the Heart. Proc Am Assoc Cancer Res. 1980; 21:266.
13. Carvalho FS, Burgeiro A, Garcia R, Moreno AJ, Carvalho RA, Oliveira PJ. Doxorubicin-induced cardiotoxicity: from bioenergetic failure and cell death to cardiomyopathy. Med Res Rev. 2014; 34(1):106–135. PMID: 23494977.
Article
14. Chia R, Achilli F, Festing MF, Fisher EM. The origins and uses of mouse outbred stocks. Nat Genet. 2005; 37(11):1181–1186. PMID: 16254564.
Article
15. Nolin TD, McMenamin ME, Himmelfarb J. Simultaneous determination of total homocysteine, cysteine, cysteinylglycine, and glutathione in human plasma by high-performance liquid chromatography: application to studies of oxidative stress. J Chromatogr B Analyt Technol Biomed Life Sci. 2007; 852(1-2):554–561.
Article
16. Myers CE, McGuire WP, Liss RH, Ifrim I, Grotzinger K, Young RC. Adriamycin: the role of lipid peroxidation in cardiac toxicity and tumor response. Science. 1977; 197(4299):165–167. PMID: 877547.
Article
17. Lu SC. Glutathione synthesis. Biochim Biophys Acta. 2013; 1830(5):3143–3153. PMID: 22995213.
Article
18. Jung YS, Kim SJ, Kwon DY, Kim YC. Comparison of the effects of buthioninesulfoximine and phorone on the metabolism of sulfur-containing amino acids in rat liver. Biochem Biophys Res Commun. 2008; 368(4):913–918. PMID: 18275846.
Article
19. Doroshow JH, Locker GY, Baldinger J, Myers CE. The effect of doxorubicin on hepatic and cardiac glutathione. Res Commun Chem Pathol Pharmacol. 1979; 26(2):285–295. PMID: 574979.
20. Indu R, Azhar TS, Nair A, Nair CK. Amelioration of doxorubicin induced cardio-and hepato-toxicity by carotenoids. J Cancer Res Ther. 2014; 10(1):62–67. PMID: 24762488.
21. Shin HJ, Cho YM, Shin HJ, Kim HD, Choi KM, Kim MG, Shin HD, Chung MW. Comparison of commonly used ICR stocks and the characterization of Korl:ICR. Lab Anim Res. 2017; 33(1):8–14. PMID: 28400834.
Article
22. Tan G, Lou Z, Liao W, Zhu Z, Dong X, Zhang W, Li W, Chai Y. Potential biomarkers in mouse myocardium of doxorubicin-induced cardiomyopathy: a metabonomic method and its application. PLoS One. 2011; 6(11):e27683. PMID: 22110719.
Article
23. Liu X, Chen Z, Chua CC, Ma YS, Youngberg GA, Hamdy R, Chua BH. Melatonin as an effective protector against doxorubicin-induced cardiotoxicity. Am J Physiol Heart Circ Physiol. 2002; 283(1):H254–H263. PMID: 12063298.
24. Fisher PW, Salloum F, Das A, Hyder H, Kukreja RC. Phosphodiesterase-5 inhibition with sildenafil attenuates cardiomyocyte apoptosis and left ventricular dysfunction in a chronic model of doxorubicin cardiotoxicity. Circulation. 2005; 111(13):1601–1610. PMID: 15811867.
Article
25. Ma Y, Zhang X, Bao H, Mi S, Cai W, Yan H, Wang Q, Wang Z, Yan J, Fan GC. Toll-like receptor (TLR) 2 and TLR4 differentially regulate doxorubicin induced cardiomyopathy in mice. PLoS One. 2012; 7(7):e40763. PMID: 22808256.
Article
26. Thayer WS. Adriamycin stimulated superoxide formation in submitochondrial particles. Chem Biol Interact. 1977; 19(3):265–278. PMID: 202411.
Article
27. Doroshow JH, Locker GY, Ifrim I, Myers CE. Prevention of doxorubicin cardiac toxicity in the mouse by N-acetylcysteine. J Clin Invest. 1981; 68(4):1053–1064. PMID: 7287901.
Article
28. Doroshow JH, Locker GY, Myers CE. Enzymatic defenses of the mouse heart against reactive oxygen metabolites: alterations produced by doxorubicin. J Clin Invest. 1980; 65(1):128–135. PMID: 7350193.
Full Text Links
  • LAR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr