Korean Circ J.  2017 Nov;47(6):960-969. 10.4070/kcj.2017.0080.

Zebrafish Larvae Model of Dilated Cardiomyopathy Induced by Terfenadine

Affiliations
  • 1Department of Microbiology and Immunology, Institute of Endemic Disease, Seoul National University College of Medicine, Seoul, Korea. hylee612@snu.ac.kr lamseok@snu.ac.kr
  • 2Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea. hylee612@snu.ac.kr lamseok@snu.ac.kr

Abstract

BACKGROUND AND OBJECTIVES
Dilated cardiomyopathy can be the end-stage of severe cardiac disorders and directly affects the cardiac muscle, inducing cardiomegaly and heart failure (HF). Although a wide variety of animal models are available to study dilated cardiomyopathy, there is no model to assess dilated cardiomyopathy with non-invasive, simple, and large screening methods.
METHODS
We developed a dilated cardiomyopathy model in zebrafish larvae using short duration terfenadine, a known cardiotoxic drug that induces ventricular size dilation. Fractional shortening of zebrafish hearts was calculated.
RESULTS
We treated zebrafish with 5 to 10 µM terfenadine for 24 hours. In terfenadine-treated zebrafish, blood frequently pooled and clotted in the chamber, and circulation was remarkably reduced. Atria and ventricles were swollen, and fluid was deposited around the heart, mimicking edema. Cardiac contractility was significantly reduced, and ventricular area was significantly enlarged. Heart rate was markedly reduced even after terfenadine withdrawal. Acridine orange staining also showed that terfenadine increased cardiomyocyte apoptosis. A significant increase of natriuretic peptide B (NPPB) mRNA was found in terfenadine-treated zebrafish. A low dose of terfenadine (5-10 µM) did not show mortality in short-term treatment (24 hours). However, moderate dose (35-45 µM) terfenadine treatment reduced zebrafish survival within 1 hour.
CONCLUSION
With advantages of rapid sample preparation procedure and transparent observation of the live heart, this model can potentially be applied to large-scale drug screening and toxicity assays for non-ischemic HF.

Keyword

Zebrafish; Heart failure; Cardiomyopathies

MeSH Terms

Acridine Orange
Apoptosis
Cardiomegaly
Cardiomyopathies
Cardiomyopathy, Dilated*
Drug Evaluation, Preclinical
Edema
Heart
Heart Failure
Heart Rate
Larva*
Mass Screening
Models, Animal
Mortality
Myocardium
Myocytes, Cardiac
RNA, Messenger
Terfenadine*
Zebrafish*
Acridine Orange
RNA, Messenger
Terfenadine

Figure

  • Figure 1 Morphology of 5 dpf zebrafish larvae after H&E staining. dpf = days post fertilization; H&E = hematoxylin and eosin.

  • Figure 2 Transient terfenadine treatment reduced heart rate, inducing blood stagnation. (A) Representative image of control zebrafish larvae treated with 0.001% DMSO for 24 hours. (B, C) Representative images of zebrafish larvae treated with 5 µM (B) and 10 µM (C) terfenadine for 24 hours. Terfenadine-treated zebrafish showed enlarged heart size (black arrow) and venous congestion (hollow arrow). (D) Heart rates in control and terfenadine-treated zebrafish larvae (n=20 zebrafish/group). DMSO = dimethyl sulfoxide. *p<0.050.

  • Figure 3 Atrioventricular dyssynchrony was induced after terfenadine treatment. (A) Representative image of cmlc-2:GFP transgenic zebrafish larvae. (B) Images taken from in vivo video recording at 4 dpf in the control (0.001% DMSO) and 24-hr terfenadine (20 μM)-treated zebrafish larvae. The interval between images in the montage is 0.196 seconds. Outlines of the atrium are shown with white dashed lines, and outlines of ventricles with red lines. (C) Arrhythmia rate of the control (0.001% DMSO) and 24-hr terfenadine (20 μM)-treated zebrafish larvae (n=20 zebrafish/group). (D) Atrial size of the control (0.001% DMSO) and 24-hr terfenadine (20 μM)-treated zebrafish larvae (n=20 zebrafish/group). A = atrium; DMSO = dimethyl sulfoxide; dpf = days post fertilization; FV/FA = fetal ventricle/fatal atrium; V = ventricle. *p<0.050.

  • Figure 4 Transient terfenadine treatment impaired cardiac contraction, resulting in HF. (A) Representative images of control (0.001% DMSO) or terfenadine (20 μM)-treated zebrafish larva. (B) Lateral view of zebrafish larvae at 4 dpf. The control zebrafish (0.001% DMSO) exhibited normal cardiac morphology, whereas terfenadine (20 μM)-treated zebrafish larvae showed pericardial edema (blue circle) and venous congestion (green circle). (C) Quantification of ventricular FS in control (0.001% DMSO) and terfenadine (20 μM)-treated zebrafish larvae. Ventricle size of control (0.001% DMSO) and terfenadine (20 μM)-treated zebrafish larvae after a 24 hours treatment. n=20 zebrafish/group, scale bar=0.5 mm. a = Atrium; DMSO = dimethyl sulfoxide; dpf = days post fertilization; FS = fractional shortening; HF = heart failure; V = ventricle. *p<0.050; †p<0.010.

  • Figure 5 Terfenadine treatment induced apoptosis in cardiomyocytes. (A) H&E staining of longitudinal sections showed normal cardiomyocyte morphology of control (0.001% DMSO) zebrafish larvae. (B) Terfenadine (20 μM)-treated zebrafish larvae for 24 hours showed elongated and thin cardiomyocytes. (C) Detection of apoptotic cardiomyocytes by acridine orange staining in the control group. (D) Detection of apoptotic cardiomyocytes by acridine orange staining in terfenadine-treated zebrafish. Outlines of ventricles are shown with red lines. Apoptotic cells are indicated by red arrows. Scale bar=0.5 μm. DMSO = dimethyl sulfoxide; dpf = days post fertilization; H&E = hematoxylin and eosin.

  • Figure 6 Terfenadine treatment induced HF and impaired survival in zebrafish larvae. (A) Quantitative RT-PCR of NPPB was increased in terfenadine-treated zebrafish larvae. (B) Survival rate of zebrafish treated with control and 35 to 45 µM of terfenadine. n=20 zebrafish/group. HF = heart failure; NPPB = natriuretic peptide B; RT-PCR = real-time polymerase chain reaction. *p<0.050.


Cited by  1 articles

A Small Animal Model of Diabetic Heart Failure With Reduced Ejection Fraction
Seung-Hyun Jung, Hyun-Taek Kim
Korean Circ J. 2023;53(1):47-48.    doi: 10.4070/kcj.2022.0349.


Reference

1. Tang C, Xie D, Feng B. Zebrafish as a new model for phenotype-based screening of positive inotropic agents. Chem Biol Drug Des. 2015; 85:253–258.
2. Huang CC, Monte A, Cook JM, Kabir MS, Peterson KP. Zebrafish heart failure models for the evaluation of chemical probes and drugs. Assay Drug Dev Technol. 2013; 11:561–572.
3. Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: the Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC)Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J. 2016; 37:2129–2200.
4. Lee SE, Cho HJ, Lee HY, et al. A multicentre cohort study of acute heart failure syndromes in Korea: rationale, design, and interim observations of the Korean Acute Heart Failure (KorAHF) registry. Eur J Heart Fail. 2014; 16:700–708.
5. Report of the WHO/ISFC task force on the definition and classification of cardiomyopathies. Br Heart J. 1980; 44:672–673.
6. Beltrami CA, Finato N, Rocco M, et al. Structural basis of end-stage failure in ischemic cardiomyopathy in humans. Circulation. 1994; 89:151–163.
7. Du CK, Morimoto S, Nishii K, et al. Knock-in mouse model of dilated cardiomyopathy caused by troponin mutation. Circ Res. 2007; 101:185–194.
8. Mahmoudabady M, Niazmand S, Shafei MN, McEntee K. Investigation of apoptosis in a canine model of chronic heart failure induced by tachycardia. Acta Physiol Hung. 2013; 100:435–444.
9. Tanaka Y, Rahmutula D, Duggirala S, et al. Diffuse fibrosis leads to a decrease in unipolar voltage: validation in a swine model of premature ventricular contraction-induced cardiomyopathy. Heart Rhythm. 2016; 13:547–554.
10. Gava FN, Zacché E, Ortiz EM, et al. Doxorubicin induced dilated cardiomyopathy in a rabbit model: an update. Res Vet Sci. 2013; 94:115–121.
11. Robert J. Long-term and short-term models for studying anthracycline cardiotoxicity and protectors. Cardiovasc Toxicol. 2007; 7:135–139.
12. Teerlink JR, Pfeffer JM, Pfeffer MA. Progressive ventricular remodeling in response to diffuse isoproterenol-induced myocardial necrosis in rats. Circ Res. 1994; 75:105–113.
13. Shih YH, Zhang Y, Ding Y, et al. Cardiac transcriptome and dilated cardiomyopathy genes in zebrafish. Circ Cardiovasc Genet. 2015; 8:261–269.
14. Bakkers J. Zebrafish as a model to study cardiac development and human cardiac disease. Cardiovasc Res. 2011; 91:279–288.
15. Jung DW, Oh ES, Park SH, et al. A novel zebrafish human tumor xenograft model validated for anti-cancer drug screening. Mol Biosyst. 2012; 8:1930–1939.
16. Asnani A, Peterson RT. The zebrafish as a tool to identify novel therapies for human cardiovascular disease. Dis Model Mech. 2014; 7:763–767.
17. Zünkler BJ, Kühne S, Rustenbeck I, Ott T. Mechanism of terfenadine block of ATP-sensitive K(+) channels. Br J Pharmacol. 2000; 130:1571–1574.
18. Hove-Madsen L, Llach A, Molina CE, et al. The proarrhythmic antihistaminic drug terfenadine increases spontaneous calcium release in human atrial myocytes. Eur J Pharmacol. 2006; 553:215–221.
19. Fearnley CJ, Roderick HL, Bootman MD. Calcium signaling in cardiac myocytes. Cold Spring Harb Perspect Biol. 2011; 3:a004242.
20. Huang CJ, Tu CT, Hsiao CD, Hsieh FJ, Tsai HJ. Germ-line transmission of a myocardium-specific GFP transgene reveals critical regulatory elements in the cardiac myosin light chain 2 promoter of zebrafish. Dev Dyn. 2003; 228:30–40.
21. Paquet D, Bhat R, Sydow A, et al. A zebrafish model of tauopathy allows in vivo imaging of neuronal cell death and drug evaluation. J Clin Invest. 2009; 119:1382–1395.
22. Milan DJ, Peterson TA, Ruskin JN, Peterson RT, MacRae CA. Drugs that induce repolarization abnormalities cause bradycardia in zebrafish. Circulation. 2003; 107:1355–1358.
23. Kooij V, Venkatraman V, Tra J, et al. Sizing up models of heart failure: proteomics from flies to humans. Proteomics Clin Appl. 2014; 8:653–664.
24. Tsang M. Zebrafish: a tool for chemical screens. Birth Defects Res C Embryo Today. 2010; 90:185–192.
25. Peterson RT, Link BA, Dowling JE, Schreiber SL. Small molecule developmental screens reveal the logic and timing of vertebrate development. Proc Natl Acad Sci USA. 2000; 97:12965–12969.
26. Schoenebeck JJ, Yelon D. Illuminating cardiac development: advances in imaging add new dimensions to the utility of zebrafish genetics. Semin Cell Dev Biol. 2007; 18:27–35.
27. Bühler A, Kustermann M, Bummer T, Rottbauer W, Sandri M, Just S. Atrogin-1 deficiency leads to myopathy and heart failure in zebrafish. Int J Mol Sci. 2016; 17:E187.
28. Cui G, Chen H, Cui W, et al. FGF2 prevents sunitinib-induced cardiotoxicity in zebrafish and cardiomyoblast H9c2 cells. Cardiovasc Toxicol. 2016; 16:46–53.
Full Text Links
  • KCJ
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