REFERENCES

1. Grunwald DJ, Eisen JS. Headwaters of the zebrafish emergence of a new model vertebrate. Nat Rev Genet 2002;3:717-24.

2. Howe K, Clark MD, Torroja CF, Torrance J, Berthelot C, Muffato M, Collins JE, Humphray S, McLaren K, Matthews L, McLaren S, Sealy I, Caccamo M, Churcher C, Scott C, Barrett JC, Koch R, Rauch GJ, White S, Chow W, Kilian B, Quintais LT, Guerra-Assunção JA, Zhou Y, Gu Y, Yen J, Vogel JH, Eyre T, Redmond S, Banerjee R, Chi J, Fu B, Langley E, Maguire SF, Laird GK, Lloyd D, Kenyon E, Donaldson S, Sehra H, Almeida-King J, Loveland J, Trevanion S, Jones M, Quail M, Willey D, Hunt A, Burton J, Sims S, McLay K, Plumb B, Davis J, Clee C, Oliver K, Clark R, Riddle C, Elliot D, Threadgold G, Harden G, Ware D, Begum S, Mortimore B, Kerry G, Heath P, Phillimore B, Tracey A, Corby N, Dunn M, Johnson C, Wood J, Clark S, Pelan S, Griffiths G, Smith M, Glithero R, Howden P, Barker N, Lloyd C, Stevens C, Harley J, Holt K, Panagiotidis G, Lovell J, Beasley H, Henderson C, Gordon D, Auger K, Wright D, Collins J, Raisen C, Dyer L, Leung K, Robertson L, Ambridge K, Leongamornlert D, McGuire S, Gilderthorp R, Griffiths C, Manthravadi D, Nichol S, Barker G, Whitehead S, Kay M, Brown J, Murnane C, Gray E, Humphries M, Sycamore N, Barker D, Saunders D, Wallis J, Babbage A, Hammond S, Mashreghi-Mohammadi M, Barr L, Martin S, Wray P, Ellington A, Matthews N, Ellwood M, Woodmansey R, Clark G, Cooper J, Tromans A, Grafham D, Skuce C, Pandian R, Andrews R, Harrison E, Kimberley A, Garnett J, Fosker N, Hall R, Garner P, Kelly D, Bird C, Palmer S, Gehring I, Berger A, Dooley CM, Ersan-Ürün Z, Eser C, Geiger H, Geisler M, Karotki L, Kirn A, Konantz J, Konantz M, Oberländer M, Rudolph-Geiger S, Teucke M, Lanz C, Raddatz G, Osoegawa K, Zhu B, Rapp A, Widaa S, Langford C, Yang F, Schuster SC, Carter NP, Harrow J, Ning Z, Herrero J, Searle SM, Enright A, Geisler R, Plasterk RH, Lee C, Westerfield M, de Jong PJ, Zon LI, Postlethwait JH, Nüsslein-Volhard C, Hubbard TJ, Roest Crollius H, Rogers J, Stemple DL. The zebrafish reference genome sequence and its relationship to the human genome. Nature 2013;496:498-503.

3. Bretaud S, Lee S, Guo S. Sensitivity of zebrafish to environmental toxins implicated in Parkinson's disease. Neurotoxicol Teratol 2004;26:857-64.

4. Hill AJ, Teraoka H, Heideman W, Peterson RE. Zebrafish as a model vertebrate for investigating chemical toxicity. Toxicol Sci 2005;86:6-19.

5. Hong RA, Iimura T, Sumida KN, Eager RM. Cardio-oncology/onco-cardiology. Clin Cardiol 2010;12:733-7.

6. Schug SA, Saunders D, Kurowski I, Paech MJ. Neuraxial drug administration: a review of treatment options for anaesthesia and analgesia. CNS Drugs 2006;20:917-33.

7. Redfern WS, Carlsson L, Davis AS, Lynch WG, MacKenzie I, Palethorpe S, Siegl PK, Strang I, Sullivan AT, Wallis R, Camm AJ, Hammond TG. Relationships between preclinical cardiac electrophysiology, clinical QT interval prolongation and torsade de pointes for a broad range of drugs: evidence for a provisional safety margin in drug development. Cardiovasc Res 2003;58:32-45.

8. Ramjaun A, AlDuhaiby E, Ahmed S, Wang L, Yu E, Nathan PC, Hodgson DC. Echocardiographic detection of cardiac dysfunction in childhood cancer survivors: how long is screening required? Pediatr Blood Cancer 2015;62:2197-203.

9. Fradley MG, Moslehi J. QT prolongation and oncology drug development. Card Electrophysiol Clin 2015;7:341-55.

10. Shah RR. Pharmacogenetic aspects of drug-induced torsade de pointes: potential tool for improving clinical drug development and prescribing. Drug Saf 2004;27:145-72.

11. Thomas D, Karle CA, Kiehn J. The cardiac hERG/IKr potassium channel as pharmacological target: structure, function, regulation, and clinical applications. Curr Pharm Des 2006;12:2271-83.

12. Biet M, Morin N, Lessard-Beaudoin M, Graham RK, Duss S, Gagné J, Sanon NT, Carmant L, Dumaine R. Prolongation of action potential duration and QT interval during epilepsy linked to increased contribution of neuronal sodium channels to cardiac late Na+ current: a potential mechanism for sudden death in epilepsy. Circ Arrhythm Electrophysiol 2015;8:912-20.

13. Langheinrich U, Vacun G, Wagner T. Zebrafish embryos express an orthologue of HERG and are sensitive toward a range of QT-prolonging drugs inducing severe arrhythmia. Toxicol Appl Pharmacol 2003;193:370-82.

14. Ducharme NA, Reif DM, Gustafsson JA, Bondesson M. Comparison of toxicity values across zebrafish early life stages and mammalian studies: implications for chemical testing. Reprod Toxicol 2014;55:3-10.

15. Lieschke GJ, Currie PD. Animal models of human disease: zebrafish swim into view. Nat Rev Genet 2007;85:353-67.

16. Wiley DS, Redfield SE, Zon LI. Chemical screening in zebrafish for novel biological and therapeutic discovery. Methods Cell Biol 2017;138:651-79.

17. Garcia GR, Noyes PD, Tanguay RL. Advancements in zebrafish applications for 21st century toxicology. Pharmacol Ther 2016;161:11-21.

18. Bartman T, Walsh EC, Wen KK, McKane M, Ren J, Alexander J, Rubenstein PA, Stainier D. Early myocardial function affects endocardial cushion development in zebrafish. PLoS Biol 2004;2:E129.

19. Poss KD, Wilson LG, Keating MT. Heart regeneration in zebrafish. Science 2002;298:2188-90.

20. Panáková D, Werdich AA, Macrae CA. Wnt11 patterns a myocardial electrical gradient through regulation of the L-type Ca(2+) channel. Nature 2010;466:874-8.

21. Andersen ND, Ramachandran KV, Bao MM, Kirby ML, Pitt GS, Hutson MR. Calcium signaling regulates ventricular hypertrophy during development independent of contraction or blood flow. J Mol Cell Cardiol 2015;80:1-9.

22. Guenancia C, Hachet O, Aboutabl M, Li N, Rigal E, Cottin Y, Rochette L, Vergely C. Overweight in mice, induced by perinatal programming, exacerbates doxorubicin and trastuzumab cardiotoxicity. Cancer Chemother Pharmacol 2016;77:777-85.

23. Vasilaki F, Tsitsimpikou C, Tsarouhas K, Germanakis I, Tzardi M, Kavvalakis M, Ozcagli E, Kouretas D, Tsatsakis AM. Cardiotoxicity in rabbits after long-term nandrolone decanoate administration. Toxicol Lett 2016;241:143-51.

24. Lamore SD, Kamendi HW, Scott CW, Dragan YP, Peters MF. Cellular impedance assays for predictive preclinical drug screening of kinase inhibitor cardiovascular toxicity. Toxicol Sci 2013;135:402-13.

25. Chu TF, Rupnick MA, Kerkela R, Dallabrida SM, Zurakowki D, Nguyen L, Woulfe K, Pravda E, Cassiola F, Desai J, George S, Morgan JA, Harris DM, Ismail NS, Chen JH, Schoen FJ, Van den Abbeele AD, Demetri GD, Force T, Chen MH. Cardiotoxicity associated with tyrosine kinase inhibitor sunitinib. Lancet 2007;370:2011-9.

26. National Research Council. Toxicity testing in the 21st century: a vision and astrategy. Washington, DC: The National Academies Press; 2007.

27. Rennekamp AJ, Peterson RT. 15 years of zebrafish chemical screening. Curr Opin Chem Biol 2015;24C:58-70.

28. Fang M, Guo J, Chen D, Li A, Hinton DE, Dong W. Halogenated carbazoles induce cardiotoxicity in developing zebrafish embryos (Danio rerio). Environ Toxicol Chem 2016;35:2523-9.

29. Cui G, Chen H, Cui W, Guo X, Fang J, Liu A, Chen Y, Lee SM. FGF2 prevents sunitinib-induced cardiotoxicity in zebrafish and cardiomyoblast H9c2 cells. Cardiovasc Toxicol 2016;16:46-53.

30. Arnaout R, Ferrer T, Huisken J, Spitzer K, Stainier DY, Tristani-Firouzi M, Chi NC. Zebrafish model for human long QT syndrome. Proc Natl Acad Sci U S A 2007;104:11316-21.

31. Sedmera D, Reckova M, deAlmeida A, Sedmerova M, Biermann M, Volejnik J, Sarre A, Raddatz E, McCarthy RA, Gourdie RG, Thompson RP. Functional and morphological evidence for a ventricular conduction system in zebrafish and Xenopus hearts. Am J Physiol Heart Circ Physiol 2003;284:H1152-60.

32. Vacaru AM, Unlu G, Spitzner M, Mione M, Knapik EW, Sadler KC. In vivo cell biology in zebrafish - providing insights into vertebrate development and disease. J Cell Sci 2014;127:485-95.

33. Stainier DY. Zebrafish genetics and vertebrate heart formation. Nat Rev Genet 2001;2:39-48.

34. Hein SJ, Lehmann LH, Kossack M, Juergensen L, Fuchs D, Katus HA, Hassel D. Advanced echocardiography in adult zebrafish reveals delayed recovery of heart function after myocardial cryoinjury. PLoS One 2015;10:e0122665.

35. Zhang CJ, Willett C, Fremgen T. Zebrafish: an animal model for toxicological studies. Curr Protoc Toxicol 2003;1:7.

36. Johnson S, Smith AG, Loffler H, Osby E, Juliusson G, Emmerich B, Wyld PJ, Hiddemann W. Multicentre prospective randomised trial of fludarabine versus cyclophosphamide, doxorubicin, and prednisone (CAP) for treatment of advanced-stage chronic lymphocytic leukaemia. The French Cooperative Group on CLL. Lancet 1996;347:1432-8.

37. Henderson IC, Berry DA, Demetri GD, Cirrincione CT, Goldstein LJ, Martino S, Ingle JN, Cooper MR, Hayes DF, Tkaczuk KH, Fleming G, Holland JF, Duggan DB, Carpenter JT, Frei E, Schilsky RL, Wood WC, Muss HB, Larry N. Improved outcomes from adding sequential paclitaxel but not from escalating doxorubicin dose in an adjuvant chemotherapy regimen for patients with node-positive primary breast cancer norton. J Clin Oncol 2003;21:976-83.

38. Peer D, Karp JM, Omid SH, Farokhzad C, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol 2007;2:751-60.

39. Woll PJ, Reichardt P, Cesne AL, Bonvalot S, Azzarelli A, Hoekstra HJ, Leahy M, Coevorden FV, Verweij J, Hogendoorn PCW, Ouali M, MSc Marreaud S, Bramwell VHC, Hohenberger P. Adjuvant chemotherapy with doxorubicin, ifosfamide, and lenograstim for resected soft-tissue sarcoma (EORTC 62931): a multicentre randomised controlled trial. Lancet Oncol 2012;13:1045-54.

40. Ling YH, el-Naggar AK, Priebe W, Perez-Soler R. Cell cycle-dependent cytotoxicity, G2/M phase arrest, and disruption of p34cdc2/cyclin B1 activity induced bydoxorubicin in synchronized P388 cells. Mol Pharmacol 1996;49:832-41.

41. Tacar O, Sriamornsak P, Dass CR. Doxorubicin: an update on anticancer molecular action, toxicity and novel drug delivery systems. J Pharm Pharmacol 2013;65:157-70.

42. Carbalho FS, Burgeiro A, Garcia R, Moreno AJ, Carbalho RA, Oliveira PJ. Doxorubicin-induced cardiotoxicity: from bioenergetic failure and cell death to cardiomyopathy. Med Res Rev 2014;34:106-35.

43. Schlitt A, Jordan K, Vordermark D, Schwamborn J, Langer T, Thomssen C. Cardiotoxicity and oncological treatments. Dtsch Arztebl Int 2014;111:161-8.

44. Völler S, Boos J, Krischke M, Würthwein G, Kontny NE, Boddy AV, Hempel G. Age-dependent pharmacokinetics of doxorubicin in children with cancer. Clin Pharmacokinet 2015;54:1139-49.

45. Zhu W, Shou W, Payne RM, Caldwell R, Field LJ. A mouse model for juvenile doxorubicin-induced cardiac dysfunction. Pediatr Res 2008;64:488-94.

46. Kim KH, Oudit GY, Backx PH. Erythropoietin protects against doxorubicin induced cardiomyopathy via a phosphatidylinositol 3-kinase-dependet pathway. J Pharmacol Exp Ther 2008;324:160-9.

47. Fischer 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;111:1601-10.

48. Delgado RM III, Nawar MA, Zewail AM, Kar B, Vaughn WK, Wu KK, Aleksic N. Sivasubramanian N, McKay K, Mann DL, Willerson JT. Cyclooxygenase-2 inhibitor treatment improves left ventricular function and mortality in a murine model of doxorubicin-induced heart failure. Circulation 2004;109:1428-33.

49. Simunek T, Striba M, Popelova O, Adamcova M, Hrdina R, Gers V. Anthracycline-induced cardiotoxicity: overview of studies examining the roles of oxidative stress and free cellular iron. Pharmacol Rep 2009;61:154-71.

50. Hassan MH, El-Beshbishy HA, Aly H, Attia SM, Bahashwan SA, Ghobara MM. Modulatory effects of meloxicam on cardiotoxicity and antitumor activity of doxorubicin in mice. Cancer Chemother Pharmacol 2014;74:559-69.

51. Gu J, Song ZP, Gui DM, Hu W, Chen YG, Zhang DD. Resveratrol attenuates doxorubicin-induced cardiomyocyte apoptosis in lymphoma nude mice by heme oxygenase-1 induction. Cardiovasc Toxicol 2012;12:341-9.

52. Chang C, Wu SL, Zhao XD, Zhao CT, Li YH. Developmental toxicity of doxorubicin hydrochloride in embryo-larval stages of zebrafish. Biomed Mater Eng 2014;24:909-16.

53. Chi NC, Shaw RM, Jungblut B, Huisken J, Ferrer T, Arnaout R, Scott I, Beis D, Xiao T, Baier H, Jan LY, Tristani-Firouzi M, Stainier DY. Genetic and physiologic dissection of the vertebrate cardiac conduction system. PLoS Biol 2008;6:e109.

54. Musso G, Tasan M, Mosimann C, Beaver JE, Plovie E, Carr LA, Chua HN, Dunham J, Zuberi K, Rodriguez H, Morris Q, Zon L, Roth FP, MacRae CA. Novel cardiovascular gene functions revealed via systematic phenotype prediction in zebrafish. Development 2014;141:224-35.

55. Rocke J, Lees J, Packham I, Chico T. The zebrafish as a novel tool for cardiovascular drug discovery. Recent Pat Cardiovasc Drug Discov 2009;4:1-5.

56. Leon C, Gerretsen P, Uchida H, Suzuki T, Rajji T, Mamo DC. Sensitivity to antipsychotic drugs in older adults. Curr Psychiatry Rep 2010;12:28-33.

57. Lasser KE, Allen PD, Woolhandler SJ, Himmelstein DU, Wolfe SM, Bor DH. Timing of new black box warnings and withdrawals for prescription medications. J Am Med Assoc 2002;287:2215-20.

58. Shah RR. Can pharmacogenetics help rescue drugs withdrawn from the market? Pharmacogenomics 2006;7:889-908.

59. Stockbridge N, Morganroth J, Shah RR, Garnett C. Dealing with global safety issues: was the response to QT-liability of non-cardiac drugs well coordinated? Drug Saf 2013;36:167-82.

60. Koponen H, Alaräisänen A, Saari K, Pelkonen O, Huikuri H, Raatikainen MJ, Savolainen M, Isohanni M. Schizophrenia and sudden cardiac death. Nord J Psychiatry 2008;62:342-5.

61. Abdel-Wahab BA, Metwally ME, El-khawanki MM, Hashim AM. Protective effect of captopril against clozapine-induced myocarditis in rats: role of oxidative stress, proinflammatory cytokines and DNA damage. Chem Biol Interact 2014;216:43-52.

62. Wang JF, Min JY, Hampton TG, Amende I, Yan X, Malek S, Abelmann WH, Green AI, Zeind J, Morgan JP. Clozapine-induced myocarditis: role of catecholamines in a murine model. Eur J Pharmacol 2008;592:123-7.

63. Dang R, Guo Y, Cai H, Yang R, Liang D, Lv C, Jiang P. Effects of prolonged antipsychotic administration on neuregulin-1/ErbB signaling in rat prefrontal cortex and myocardium: implications for the therapeutic action and cardiac adverse effect. J Toxicol Sci 2016;41:303-9.

64. Barbazuk WB, Korf I, Kadavi C, Heyen J, Tate S, Wun E, Bedell JA, McPherson JD, Johnson SL. The syntenic relationship of the zebrafish and human genomes. Genome Res 2000;10:1351-8.

65. Lee SH, Kim HR, Han RX, Oqani RK, Jin DI. Cardiovascular risk assessment of atypical antipsychotic drugs in a zebrafish model. J Appl Toxicol 2013;33:466-70.

66. Pylatiuk C, Sanchez D, Mikut R, Alshut R, Reischl M, Hirth S, Rottbauer W, Just S. Automatic zebrafish heartbeat detection and analysis for zebrafish embryos. Zebrafish 2014;11:379-83.

67. Manjanatha MG, Bishop ME, Pearce MG, Kulkarni R, Lyn-Cook LE, Ding W. Genotoxicity of doxorubicin in F344 rats by combining the comet assay, flow-cytometric peripheral blood micronucleus test, and pathway-focused gene expression profiling. Environ Mol Mutagen 2014;55:24-34.

68. Faßbender C, Braunbeck T. Assessment of genotoxicity in gonads, liver and gills of zebrafish (Danio rerio) by use of the comet assay and micronucleus test after in vivo exposure to methyl methanesulfonate. Bull Environ Contam Toxicol 2013;91:89-95.

69. Kovács R, Csenki Z, Bakos K, Urbányi B, Horváth Á, Garaj-Vrhovac V, Gajski G, Gerić M, Negreira N, López de Alda M, Barceló D, Heath E, Kosjek T, Žegura B, Novak M, Zajc I, Baebler Š, Rotter A, Ramšak Ž, Filipič M. Assessment of toxicity and genotoxicity of low doses of 5-fluorouracil in zebrafish (Danio rerio) two-generation study. Water Res 2015;77:201-12.

70. Devaux A, Pesonen M, Monod G. Alkaline comet assay in rainbow trout hepatocytes. Toxicol In Vitro 1997;11:71-9.

71. Šrut M, Traven L, Štambuk A, Kralj S, Žaja R, Mićović V, Klobučar GIV. Genotoxicity of marine sediments in the fish hepatoma cell line PLHC-1 as assessed by the Comet assay. Toxicol In Vitro 2011;25:308-14.

72. Šrut M, Bourdineaud JP, Štambuk A, Klobučar GI. Genomic and gene expression responses to genotoxic stress in PAC2 zebrafish embryonic cell line. J Appl Toxicol 2015;35:1381-9.

73. Mladěnka P, Applová L, Patočka J, Costa VM, Remiao F, Pourová J, Mladěnka A, Karlíčková J, Jahodář L, Vopršalová M, Varner KJ, Štěrba M; TOX-OER and CARDIOTOX Hradec Králové Researchers and Collaborators. Comprehensive review of cardiovascular toxicity of drugs and related agents. Med Res Rev 2018; doi: 10.1002/med.21476.

74. Chan J, Bayliss PE, Wood JM, Roberts TM. Dissection of angiogenic signaling in zebrafish using a chemical genetic approach. Cancer Cell 2002;1:257-67.

75. Jing L, Durand EM, Ezzio C, Pagliuca SM, Zon LI. In situ hybridization assay-based small molecule screening in zebrafish. Curr Protoc Chem Biol 2012;4:110236.

76. Lancaster MC, Sobie EA. Improved prediction of drug-induced Torsades de Pointes through simulations of dynamics and machine learning algorithms. Clin Pharmacol Ther 2016;100:371-9.

77. Glinka A, Polak S. The effects of six antipsychotic agents on QTc an attempt to mimic clinical trial through simulation including variability in the population. Comput Biol Med 2014;47:20-6.

78. Holmgren G, Synnergren J, Bogestål Y, Améen C, Åkesson K, Holmgren S, Lindahl A, Sartipy P. Identification of novel biomarkers for doxorubicin-induced toxicity in human cardiomyocytes derived from pluripotent stem cells. Toxicology 2015;328:102-11.

79. Yuan Y, Bai X, Luo C, Wang K, Zhang H. The virtual heart as a platform for screening drug cardiotoxicity. Br J Pharmacol 2014;172:5531-47.

80. Pott A, Rottbauer W, Just S. Functional genomics in zebrafish as a tool to identify novel antiarrhythmic targets. Curr Med Chem 2014;21:1320-9.

81. Mishra S, Guan J, Plovie E, Seldin DC, Connors LH, Merlini G, Falk RH, MacRae CA, Liao R. Human amyloidogenic light chain proteins result in cardiac dysfunction, cell death, and early mortality in zebrafish. Am J Physiol 2013;305:H95-103.

82. Hwang WY, Fu Y, Reyon D, Maeder ML, Tsai SQ, Sander JD, Peterson RT, Yeh JR, Joung JK. Efficient genome editing in zebrafish using a CRISPR-Cas system. Nat Biotechnol 2013;31:227-31.

83. Bedell VM, Wang Y, Campbell JM, Poshusta TL, Starker CG, Krug RG, Tan W, Penheiter SG, Ma AC, Leung AY, Fahrenkrug SC, Carlson DF, Voytas DF, Clark KJ, Essner JJ, Ekker SC. In vivo genome editing using a high-efficiency TALEN system. Nature 2012;491:114-8.

84. Doyon Y, McCammon JM, Miller JC, Faraji F, Ngo C, Katibah GE, Amora R, Hocking TD, Zhang L, Rebar EJ, Gregory PD, Urnov FD, Amacher SL. Heritable targeted gene disruption in zebrafish using designed zinc-finger nucleases. Nat Biotechnol 2008;26:702-8.

85. Marelli F, Persani L. How zebrafish research has helped in understanding thyroid diseases. F1000Res 2017;6:2137.

86. Zon LI, Peterson RT. In vivo drug discovery in the zebrafish. Nat Rev Drug Discov 2005;4:35-44.

87. Dinday MT, Baraban SC. Large-scale phenotype-based antiepileptic drug screening in a zebrafish model of dravet syndrome. eNeuro 2015;2:ENEURO.0068-15.2015.

88. Dravet C, Bureau M, Oguni H, Fukuyama Y, Cokar O. Severe myoclonic epilepsy in infancy: Dravet syndrome. Adv Neurol 2005;95:71-102.

89. Ceulemans B, Boel M, Leyssens K, Van Rossem C, Neels P, Jorens PG, Lagae L. Successful use of fenfluramine as an add-on treatment for Dravet syndrome. Epilepsia 2012;53:1131-9.

90. Stainier DY, Fouquet B, Chen JN, Warren KS, Weinstein BM, Meiler SE, Mohideen MA, Neuhauss SC, Solnica-Krezel L, Schier AF, Zwartkruis F, Stemple DL, Malicki J, Driever W, Fishman MC. Mutations affecting the formation and function of the cardiovascular system in the zebrafish embryo. Development 1996;123:285-92.

91. Knöll R, Postel R, Wang J, Krätzner R, Hennecke G, Vacaru AM, Vakeel P, Schubert C, Murthy K, Rana BK, Kube D, Knöll G, Schäfer K, Hayashi T, Holm T, Kimura A, Schork N, Toliat MR, Nürnberg P, Schultheiss HP, Schaper W, Schaper J, Bos E, Den Hertog J, van Eeden FJ, Peters PJ, Hasenfuss G, Chien KR, Bakkers J. Laminin-alpha4 and integrin-linked kinase mutations cause human cardiomyopathy via simultaneous defects in cardiomyocytes and endothelial cells. Circulation 2007;116:515-25.

92. Kettleborough RN, Busch-Nentwich EM, Harvey SA, Dooley CM, de Bruijn E, van Eeden F, Sealy I, White RJ, Herd C, Nijman IJ, Fényes F, Mehroke S, Scahill C, Gibbons R, Wali N, Carruthers S, Hall A, Yen J, Cuppen E, Stemple DL. A systematic genome-wide analysis of zebrafish protein-coding gene function. Nature 2013;496:494-7.

93. Zhu XW, Li SJ. In silico prediction of drug-induced liver injury based on adverse drug reaction reports. Toxicol Sci 2017;158:391-400.

94. Sewing S, Boess F, Moisan A, Bertinetti-Lapatki C, Minz T, Hedtjaern M, Tessier Y, Schuler F, Singer T, Roth AB. Establishment of a predictive in vitro assay for assessment of the hepatotoxic potential of oligonucleotide drugs. PLoS One 2016;11:e0159431.

95. Kleiner DE. Drug-induced liver injury: the hepatic pathologist's approach. Gastroenterol Clin North Am 2017;46:273-96.

96. Chu J, Kirsten C, Sadler A. New school in liver development: lessons from zebrafish. Hepatology 2009;50:1656-63.

97. Menke AL, Spitsbergen JM, Wolterbeek AP, Woutersen RA. Normal anatomy and histology of the adult zebrafish. Toxicol Pathol 2011;39:759-75.

98. Quinlivan VH, Farber SA. Lipid uptake, metabolism, and transport in the larval zebrafish. Front Endocrinol (Lausanne) 2017;8:319.

99. Vliegenthart AD, Tucker CS, Del Pozo J, Dear JW. Zebrafish as model organisms for studying drug-induced liver injury. Br J Clin Pharmacol 2014;78:1217-27.

100. Zhang X, Li C, Gong Z. Development of a convenient in vivo hepatotoxin assay using a transgenic zebrafish line with liver-specific dsred expression. PLoS One 2014;9:e91874.

101. Saad M, Matheeussen A, Bijttebier S, Verbueken E, Pype C, Casteleyn C, Van Ginneken C, Apers S, Maes L, Cos P, Van Cruchten S. In vitro CYP-mediated drug metabolism in the zebrafish (embryo) using human reference compounds. Toxicol In Vitro 2017;42:329-36.

102. Mesens N, Crawford AD, Menke A, Hung PD, Van Goethem F, Nuyts R, Hansen E, Wolterbeek A, Van Gompel J, De Witte P, Esguerra CV. Are zebrafish larvae suitable for assessing the hepatotoxicity potential of drug candidates? J Appl Toxicol 2015;35:1017-29.

103. Cornet C, Calzolari S, Miñana-Prieto R, Dyballa S, van Doornmalen E, Rutjes H, Savy T, D'Amico D, Terriente J. ZeGlobalTox: an innovative approach to address organ drug toxicity using zebrafish. Int J Mol Sci 2017;18:E864.

104. Nahar K, Gupta N, Gauvin R, Absar S, Patel B, Gupta V, Khademhosseini A, Ahsan F. In vitro, in vivo and ex vivo models for studying particle deposition and drug absorption of inhaled pharmaceuticals. Eur J Pharm Sci 2013;49:805-18.

105. Farghali H, Kgalalelo Kemelo M, Wojnarová L, Kutinová Canová N. In vitro and in vivo experimental hepatotoxic models in liver research: applications to the assessment of potential hepatoprotective drugs. Physiol Res 2016;65:S417-25.

106. Schaeck M, Van den Broeck W, Hermans K, Decostere A. Fish as research tools: alternatives to in vivo experiments. Altern Lab Anim 2013;41:219-29.

Journal of Unexplored Medical Data
ISSN 2572-8180 (Online)
Follow Us

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/