Open Access Open Access  Restricted Access Subscription Access

Fish models in experimental pharmacology: on the mark or off the mark?


Affiliations
1 Department of Pharmacology, College of Pharmaceutical Sciences, Government Medical College, Thiruvananthapuram 695 011, India, India
2 Marine Biotechnology Division, ICAR-Central Marine Fisheries Research Institute, Kochi 682 018, India, India
3 ICAR-Central Coastal Agricultural Research Institute, Old Goa, Goa 403 402, India, India
 

Fish has emerged as an alternative model organism in biomedical research for conducting experimental pharmacological and toxicological studies. As a vertebrate, it shares many conserved physiological and molecular features with humans making it a valuable model for diagnosing, investigating disease states and testing drugs to check toxicity and therapeutic activity against the target. Zebrafish and medaka are mainstream models that are widely employed in pharmaceutical research. This study aims to highlight the probability and potential of fish as an alternative model organism in biomedical research, drug discovery and development. Further, it discusses the limitations of fish models in experimental pharmacological and toxicological studies considering the changes in the residing environment, physiology, metabolism, unpredictable inter-individual variability due to diseases, variable conditioning, and interspecific and intraspecific variability

Keywords

Drug screening, fish, model organism, phar-macology, toxicology.
User
Notifications
Font Size

  • Bolis, C. L., Piccolella, M., Dalla Valle, A. Z. and Rankin, J. C., Fish as model in pharmacological and biological research. Pharma-col. Res., 2001, 44, 265–280.
  • Schartl, M., Beyond the zebrafish: diverse fish species for model-ing human disease. Dis. Models Mech., 2014, 7, 181–192.
  • Schaeck, M., Broeck, W. V. D., Hermans, K. and Decostere, A., Fish as research tools: alternatives to in vivo experiments. Altern. Lab. Anim., 2013, 41, 219–229.
  • Teame, T. et al., The use of zebrafish (Danio rerio) as biomedical models. Anim. Front., 2019, 9, 68–77.
  • Hilgers, L. and Schwarzer, J., The natural history of model organ-isms: the untapped potential of medaka and its wild relatives. eLife, 2019, 8, e46994.
  • Simonetti, R. B., Marques, L. S., Streit, D. P. and Oberst, E. R., Zebrafish (Danio rerio): the future of animal model in biomedical research. J. FishSci.com, 2015, 9, 39–45.
  • Pandey, G., A review of fish model in experimental pharmacology. Int. Res. J. Pharm., 2011, 2, 33–36.
  • Law, J. M., Mechanistic considerations in small fish carcinogenicity testing. Inst. Lab. Anim. Res. J., 2001, 42, 274–284.
  • Bunton, T. E., Experimental chemical carcinogenesis in fish. Toxi-col. Pathol., 1996, 24, 603–618.
  • van de Pol, I., Flik, G. and Gorrisen, M., Comparative physiology of energy metabolism: fishing for endocrine signals in the early vertebrate pool. Front. Endocrinol., 2017, 8, 36.
  • Lin, C. Y., Chiang, C. Y. and Tsai, H. J., Zebrafish and medaka: new model organisms for modern biomedical research. J. Biomed. Sci., 2016, 23, 19.
  • Toutain, P. L., Ferran, A. and Melou, A. B., Species differences in pharmacokinetics and pharmacodynamics. Handb. Exp. Pharma-col., 2010, 199, 19–48.
  • Strange, K., Drug discovery in fish, flies, and worms. Inst. Lab. Anim. Res. J., 2016, 57, 133–143.
  • Szabo, M. et al., Cell and small animal models for phenotypic drug discovery. Drug Des. Dev. Ther., 2017, 11, 1957–1967.
  • MacRae, C. A. and Peterson, R. T., Zebrafish as tools for drug dis-covery. Nature Rev., 2015, 14, 721–731.
  • Giacomotto, J. and Ségalat, L., High-throughput screening and small animal models, where are we? Br. J. Pharmacol., 2010, 160, 204–216.
  • Caballero, M. V. and Candiracci, M., Zebrafish as screening model for detecting toxicity and drug’s efficacy. J. Unexplor. Med. Data, 2018, 3, 4.
  • Chitmanat, C., Tongdonmuan, K. and Nunsong, W., The use of crude extracts from traditional medicinal plants to eliminate Trichodina sp. in tilapia (Oreochromis niloticus) fingerlings. Songklanakarin J. Sci. Technol., 2005, 27, 359–364.
  • Dugenci, S. K., Arda, N. and Candan, A., Some medicinal plants as immunostimulant for fish. J. Ethnopharmacol., 2003, 88, 99– 106.
  • Winkaler, E. U., Santos, T. R. M., Machado-Neto, J. G. and Mar-tinez, C. B. R., Acute lethal and sublethal effects of neem leaf ex-tract on the neotropical freshwater fish Prochilodus lineatus. Comp. Biochem. Physiol. Part C, 2007, 145, 236–244.
  • Atala, A., Regenerative medicine strategies. J. Pediatr. Surg., 2012, 47, 17–28.
  • Gemberling, M., Bailey, T. J., Hyde, D. R. and Poss, K. D., The zebrafish as a model for complex tissue regeneration. Trends Genet., 2013, 29, 1–19.
  • Marques, I. J., Lupi, E. and Mercader, N., Model systems for regeneration: zebrafish. Development, 2019, 146, 1–13.
  • Pfefferli, C. and Jazwinska, A., The art of fin regeneration in zebra-fish. Regeneration, 2015, 2, 72–83.
  • Chassot, B., Pury, D. and Jaźwińska, A., Zebrafish fin regeneration after cryoinjury-induced tissue damage. Biol. Open, 2016, 5, 819– 828.
  • Yu, F., Li, R., Parks, E., Takabe, W. and Hsiai, T. K., Electrocardi-ogram signals to assess zebrafish heart regeneration: implication of long QT intervals. Annu. Rev. Biomed. Eng., 2010, 38, 2346–2357.
  • Wang, J. et al., The regenerative capacity of zebrafish reverses car-diac failure caused by genetic cardiomyocyte depletion. Develop-ment, 2011, 138, 3421–3430.
  • Chablais, F., Veit, J., Rainer, G. and Jaźwińska, A., The zebrafish heart regenerates after cryoinjury-induced myocardial infarction. BMC Dev. Biol., 2011, 11, 21.
  • Reimschuessel, R. A., Fish model of renal regeneration and devel-opment. ILAR, 2001, 42, 285–291.
  • Goldshmit, Y., Sztal, T. E., Jusuf, P. R., Hall, T. E., Nguyen-Chi, M. and Currie, P. D., Fgf-dependent glial cell bridges facilitate spi-nal cord regeneration in zebrafish. Neuroscience, 2012, 32, 7477– 7492.
  • Marz, M. et al., Regenerative response following stab injury in the adult zebrafish telencephalon. Dev. Dyn., 2011, 240, 2221–2231.
  • Diekmann, H., Kalbhen, P. and Fischer, D., Characterization of optic nerve regeneration using transgenic zebrafish. Front. Cell. Neurosci., 2015, 9, 118.
  • North, T. E. et al., PGE2-regulated Wnt signaling and N-acetyl-cysteine are synergistically hepatoprotective in zebrafish aceta-minophen injury. Proc. Natl. Acad. Sci. USA, 2010, 107, 17315– 17320.
  • Kan, N. G., Junghans, D., Izpisua and Belmonte, J. C., Compensa-tory growth mechanisms regulated by BMP and FGF signaling me-diate liver regeneration in zebrafish after partial hepatectomy. FASEB J., 2009, 23, 3516–3525.
  • Ye, L., Robertson, M. A., Mastracci, T. L. and Anderson, R. M., An insulin signaling feedback loop regulates pancreas progenitor cell differentiation during islet development and regeneration. Dev. Biol., 2016, 409, 354–369.
  • Richardson, R. et al., Adult zebrafish as a model system for cutane-ous wound-healing research. J. Invest. Dermatol., 2013, 133, 1665.
  • Unguez, G. A., Electric fish: new insights into conserved processes of adult tissue regeneration. J. Exp. Biol., 2013, 216, 2478–2486.
  • Bhat, S. A., Scenario of genotoxicity in fishes and its impact on fish industry. IOSR-JESTFT, 2014, 8, 65–76.
  • Banu, B. S., Danadevi, K., Rahman, M. F., Ahuja, Y. R. and Kai-ser, J., Genotoxic effect of monocrotophos to sentinel species using comet assay. Food Chem. Toxicol., 2001, 39, 361–366.
  • D’Costa, A. H., Shyama, S. K., Kumar, M. K. P. and Fernandes, T. M., Induction of DNA damage in the peripheral blood of zebrafish (Danio rerio) by an agricultural organophosphate pesticide, mono-crotophos. Int. Aquat. Res., 2018, 10, 243–251.
  • Dash, M. R. and Soren, D., Testing of genotoxic potential of ami-kacin sulphate through micronucleus test in a fish in vivo system. Int. Res. J. Biol. Sci., 2018, 7, 36–40.
  • Obiakor, M. O., Okonkwo, J. C. and Ezeonyejiaku, C. D., Genotoxi-city of freshwater ecosystem shows DNA damage in preponderant fish as validated by in vivo micronucleus induction in gill and kid-ney erythrocytes. Mutat. Res.-Genet. Toxicol. Environ. Mutagen., 2014, 775–776, 20–30.
  • Francisco, C. D. M., Bertolino, S. M., De Oliveira, R. J., Morelli, S. and Pereira, B. B., Genotoxicity assessment of polluted urban streams using a native fish Astyanax altiparanae. J. Toxicol. Envi-ron. Health, 82, 514–523.
  • Li, H. and Zhang, S., In vitro cytotoxicity of the organophosphorus pesticide parathion to FG-9307 cells. Toxicol. Vitro, 2001, 15, 643– 647.
  • Patton, E. E., Zon, L. I. and Langenau, D. M., Zebrafish disease models in drug discovery: from preclinical modelling to clinical trials. Nature Rev. Drug Discov., 2021, 20, 611–628.
  • Santoriello, C. and Zon, L. I., Hooked! Modeling human disease in zebrafish. J. Clin. Invest., 2012, 122, 2337–2343.
  • Meshalkina, D. A., Kysil, E. V., Warnick, J. E., Demin, K. A. and Kaluef, A. V., Adult zebrafish in CNS disease modeling: a tank that’s half-full, not half-empty, and still filling. Lab. Anim., 2017, 46, 378–387.
  • Matsumoto, T., Terai, S. and Oishi, T., Medaka as a model for human nonalcoholic steatohepatitis. Dis. Models Mech., 2010, 3, 431–440.
  • Matsui, H., Gavinio, R. and Takahashi, R., Medaka fish Parkin-son’s disease model. Exp. Neurobiol., 2012, 21, 94–100.
  • Walter, R. B. and Obara, T., Workshop report: the medaka model for comparative assessment of human disease mechanisms. Comp. Biochem. Physiol. Part C, 2015, 178, 156–162.
  • Usai, A. et al., A model of a zebrafish avatar for co-clinical trials. Cancers (Basel), 2020, 12, 677.
  • Precazzini, F. et al., Automated in vivo screen in zebrafish identi-fies clotrimazole as targeting a metabolic vulnerability in a mela-noma model. Dev. Biol., 2020, 457, 215–225.
  • Stewart, A. M. et al., Molecular psychiatry of zebrafish. Mol. Psy-chiatry, 2015, 20, 2–17.
  • Ota, K. G. and Abe, G., Goldfish morphology as a model for evolu-tionary developmental biology. WIREs Dev. Biol., 2016, 5, 272–295.
  • Kondo, M., Nanda, I., Schmid, M. and Schartl, M., Sex determina-tion and sex chromosome evolution: insights from medaka. Sex. Dev., 2009, 3, 88–98.
  • Herrera, M. and Jagadeeswaran, P., Annual fish as a genetic model for aging. Biol. Sci., 2004, 59, 101–107.
  • Hoo, J. Y., Kumari, Y., Shaikh, M. F., Hue, S. M. and Goh, B. H., Zebrafish: a versatile animal model for fertility research. BioMed. Res. Int., 2016, 2016, 9732780.
  • Bradford, Y. M. et al., Zebrafish models of human disease: gaining insight into human disease at ZFIN. Inst. Lab. Anim. Res. J., 2017, 58, 4–16.
  • Li, S. et al., Constructing a fish metabolic network model. Genome Biol., 2010, 11, 115.
  • Wang, N. et al., Exploration of age-related mitochondrial dysfunc-tion and the anti-aging effects of resveratrol in zebrafish retina. Ag-ing, 2019, 11, 3117–3137.
  • Dugershaw, B. B., Aengenheister, L., Hansen, S. S. K., Hougaard, K. S. and Thurnherr, T. B., Recent insights on indirect mechanisms in developmental toxicity of nanomaterials. Part. Fibre Toxicol., 2020, 17, 31.
  • Lawrence, C. et al., Regulatory compliance and the zebrafish. Zebrafish, 2009, 6, 453–456.
  • DeTolla, L. J. et al., Guidelines for the care and use of fish in re-search. Inst. Lab. Anim. Res. J., 1995, 37, 159–173.
  • Malaga-trillo, E., Laessing, U., Lang, D. M. and Stuermer, C. A. O., Evolution of duplicated reggie genes in zebrafish and goldfish. J. Mol. Evol., 2002, 54, 235–245.
  • Lee, H. C., Lin, C. Y. and Tsai, H. J., Zebrafish, an in vivo platform to screen drugs and proteins for biomedical use. Pharmaceuticals, 2021, 14, 500.

Abstract Views: 138

PDF Views: 80




  • Fish models in experimental pharmacology: on the mark or off the mark?

Abstract Views: 138  |  PDF Views: 80

Authors

Supriya Raja Harikumar
Department of Pharmacology, College of Pharmaceutical Sciences, Government Medical College, Thiruvananthapuram 695 011, India, India
Reshma Janardhanan
Marine Biotechnology Division, ICAR-Central Marine Fisheries Research Institute, Kochi 682 018, India, India
Giri Bhavan Sreekanth
ICAR-Central Coastal Agricultural Research Institute, Old Goa, Goa 403 402, India, India

Abstract


Fish has emerged as an alternative model organism in biomedical research for conducting experimental pharmacological and toxicological studies. As a vertebrate, it shares many conserved physiological and molecular features with humans making it a valuable model for diagnosing, investigating disease states and testing drugs to check toxicity and therapeutic activity against the target. Zebrafish and medaka are mainstream models that are widely employed in pharmaceutical research. This study aims to highlight the probability and potential of fish as an alternative model organism in biomedical research, drug discovery and development. Further, it discusses the limitations of fish models in experimental pharmacological and toxicological studies considering the changes in the residing environment, physiology, metabolism, unpredictable inter-individual variability due to diseases, variable conditioning, and interspecific and intraspecific variability

Keywords


Drug screening, fish, model organism, phar-macology, toxicology.

References





DOI: https://doi.org/10.18520/cs%2Fv123%2Fi10%2F1199-1206