[1] LEGRADI J B, Di PAOLO C, KRAAK M H S, et al. An ecotoxicological view on neurotoxicity assessment [J]. Environmental Sciences Europe, 2018, 30(1): 46. doi: 10.1186/s12302-018-0173-x
[2] WLODKOWIC D, CAMPANA O. Toward high-throughput fish embryo toxicity tests in aquatic toxicology [J]. Environmental Science & Technology, 2021, 55(6): 3505-3513.
[3] KAVLOCK R, CHANDLER K, HOUCK K, et al. Update on EPA’s ToxCast program: Providing high throughput decision support tools for chemical risk management [J]. Chemical Research in Toxicology, 2012, 25(7): 1287-1302. doi: 10.1021/tx3000939
[4] CAMPANA O, WLODKOWIC D. Ecotoxicology goes on a chip: Embracing miniaturized bioanalysis in aquatic risk assessment [J]. Environmental Science & Technology, 2018, 52(3): 932-946.
[5] KREWSKI D, ACOSTA D, ANDERSEN M, et al. Toxicity testing in the 21st century: A vision and a strategy [J]. Journal of Toxicology and Environmental Health, Part B, 2010, 13(2/3/4): 51-138.
[6] BUGEL S M, TANGUAY R L, PLANCHART A. Zebrafish: A marvel of high-throughput biology for 21st century toxicology [J]. Current Environmental Health Reports, 2014, 1(4): 341-352. doi: 10.1007/s40572-014-0029-5
[7] GARCIA G R, NOYES P D, TANGUAY R L. Advancements in zebrafish applications for 21st century toxicology [J]. Pharmacology & Therapeutics, 2016, 161: 11-21.
[8] DRIEVER W, SOLNICA-KREZEL L, SCHIER A F, et al. A genetic screen for mutations affecting embryogenesis in zebrafish [J]. Development (Cambridge, England), 1996, 123: 37-46. doi: 10.1242/dev.123.1.37
[9] HAFFTER P, GRANATO M, BRAND M, et al. The identification of genes with unique and essential functions in the development of the zebrafish, Danio rerio [J]. Development (Cambridge, England), 1996, 123: 1-36. doi: 10.1242/dev.123.1.1
[10] EISEN J S. History of zebrafish research[M]//The Zebrafish in Biomedical Research. Amsterdam: Elsevier, 2020: 3-14.
[11] LAM S H, WU Y L, VEGA V B, et al. Conservation of gene expression signatures between zebrafish and human liver tumors and tumor progression [J]. Nature Biotechnology, 2006, 24(1): 73-75. doi: 10.1038/nbt1169
[12] HOWE K, CLARK M D, TORROJA C F, et al. The zebrafish reference genome sequence and its relationship to the human genome [J]. Nature, 2013, 496(7446): 498-503. doi: 10.1038/nature12111
[13] PHILLIPS J B, WESTERFIELD M. Zebrafish models in translational research: Tipping the scales toward advancements in human health [J]. Disease Models & Mechanisms, 2014, 7(7): 739-743.
[14] PATTON E E, ZON L I, LANGENAU D M. Zebrafish disease models in drug discovery: From preclinical modelling to clinical trials [J]. Nature Reviews Drug Discovery, 2021, 20(8): 611-628. doi: 10.1038/s41573-021-00210-8
[15] MacRAE C A, PETERSON R T. Zebrafish as tools for drug discovery [J]. Nature Reviews Drug Discovery, 2015, 14(10): 721-731. doi: 10.1038/nrd4627
[16] PETERSON R T, LINK B A, DOWLING J E, et al. Small molecule developmental screens reveal the logic and timing of vertebrate development [J]. Proceedings of the National Academy of Sciences of the United States of America, 2000, 97(24): 12965-12969. doi: 10.1073/pnas.97.24.12965
[17] CHOE C P, CHOI S Y, YUN K E, et al. Transgenic fluorescent zebrafish lines that have revolutionized biomedical research [J]. Laboratory Animal Research, 2021, 37(1): 26. doi: 10.1186/s42826-021-00103-2
[18] RENNEKAMP A J, PETERSON R T. 15 years of zebrafish chemical screening [J]. Current Opinion in Chemical Biology, 2015, 24: 58-70. doi: 10.1016/j.cbpa.2014.10.025
[19] VOLZ D C, HIPSZER R A, LEET J K. Leveraging embryonic zebrafish to prioritize ToxCast testing [J]. Environmental Science & Technology Letters, 2015, 2(7): 171-176.
[20] NISHIMURA Y, INOUE A, SASAGAWA S, et al. Using zebrafish in systems toxicology for developmental toxicity testing [J]. Congenital Anomalies, 2016, 56(1): 18-27. doi: 10.1111/cga.12142
[21] CASSAR S, ADATTO I, FREEMAN J L, et al. Use of zebrafish in drug discovery toxicology [J]. Chemical Research in Toxicology, 2020, 33(1): 95-118. doi: 10.1021/acs.chemrestox.9b00335
[22] LIN S J, ZHAO Y, NEL A E, et al. Zebrafish: An in vivo model for nano EHS studies [J]. Small, 2013, 9(9/10): 1608-1618.
[23] GUSTAFSON A L, STEDMAN D B, BALL J, et al. Inter-laboratory assessment of a harmonized zebrafish developmental toxicology assay–Progress report on phase I [J]. Reproductive Toxicology, 2012, 33(2): 155-164. doi: 10.1016/j.reprotox.2011.12.004
[24] BRANNEN K C, PANZICA-KELLY J M, DANBERRY T L, et al. Development of a zebrafish embryo teratogenicity assay and quantitative prediction model [J]. Birth Defects Research. Part B, Developmental and Reproductive Toxicology, 2010, 89(1): 66-77. doi: 10.1002/bdrb.20223
[25] PADILLA S, CORUM D, PADNOS B, et al. Zebrafish developmental screening of the ToxCast™ Phase I chemical library [J]. Reproductive Toxicology, 2012, 33(2): 174-187. doi: 10.1016/j.reprotox.2011.10.018
[26] TRUONG L, REIF D M, ST MARY L, et al. Multidimensional in vivo hazard assessment using zebrafish [J]. Toxicological Sciences, 2014, 137(1): 212-233. doi: 10.1093/toxsci/kft235
[27] van den BULCK K, HILL A, MESENS N, et al. Zebrafish developmental toxicity assay: A fishy solution to reproductive toxicity screening, or just a red herring? [J]. Reproductive Toxicology, 2011, 32(2): 213-219. doi: 10.1016/j.reprotox.2011.06.119
[28] SELDERSLAGHS I W T, BLUST R, WITTERS H E. Feasibility study of the zebrafish assay as an alternative method to screen for developmental toxicity and embryotoxicity using a training set of 27 compounds [J]. Reproductive Toxicology, 2012, 33(2): 142-154. doi: 10.1016/j.reprotox.2011.08.003
[29] PANZICA-KELLY J M, ZHANG C X, DANBERRY T L, et al. Morphological score assignment guidelines for the dechorionated zebrafish teratogenicity assay [J]. Birth Defects Research. Part B, Developmental and Reproductive Toxicology, 2010, 89(5): 382-395. doi: 10.1002/bdrb.20260
[30] SIPES N S, PADILLA S, KNUDSEN T B. Zebrafish: As an integrative model for twenty-first century toxicity testing [J]. Birth Defects Research. Part C, Embryo Today:Reviews, 2011, 93(3): 256-267. doi: 10.1002/bdrc.20214
[31] HURTT M E, CAPPON G D, BROWNING A. Proposal for a tiered approach to developmental toxicity testing for veterinary pharmaceutical products for food-producing animals [J]. Food and Chemical Toxicology, 2003, 41(5): 611-619. doi: 10.1016/S0278-6915(02)00326-5
[32] OLSON H, BETTON G, ROBINSON D, et al. Concordance of the toxicity of pharmaceuticals in humans and in animals [J]. Regulatory Toxicology and Pharmacology, 2000, 32(1): 56-67. doi: 10.1006/rtph.2000.1399
[33] PANZICA-KELLY J M, ZHANG C X, AUGUSTINE-RAUCH K A. Optimization and performance assessment of the chorion-off[dechorinated]zebrafish developmental toxicity assay [J]. Toxicological Sciences, 2015, 146(1): 127-134. doi: 10.1093/toxsci/kfv076
[34] von HELLFELD R, BROTZMANN K, BAUMANN L, et al. Adverse effects in the fish embryo acute toxicity (FET) test: A catalogue of unspecific morphological changes versus more specific effects in zebrafish (Danio rerio) embryos [J]. Environmental Sciences Europe, 2020, 32(1): 122. doi: 10.1186/s12302-020-00398-3
[35] HAMM J T, CEGER P, ALLEN D, et al. Characterizing sources of variability in zebrafish embryo screening protocols [J]. ALTEX, 2019, 36(1): 103-120. doi: 10.14573/altex.1804162
[36] LI S Y, JIANG Y, SUN Q Q, et al. Tebuconazole induced oxidative stress related hepatotoxicity in adult and larval zebrafish (Danio rerio) [J]. Chemosphere, 2020, 241: 125129. doi: 10.1016/j.chemosphere.2019.125129
[37] MOSER V C. Functional assays for neurotoxicity testing [J]. Toxicologic Pathology, 2011, 39(1): 36-45. doi: 10.1177/0192623310385255
[38] ORGER M B, de POLAVIEJA G G. Zebrafish behavior: Opportunities and challenges [J]. Annual Review of Neuroscience, 2017, 40: 125-147. doi: 10.1146/annurev-neuro-071714-033857
[39] KALUEFF A V, GEBHARDT M, STEWART A M, et al. Towards a comprehensive catalog of zebrafish behavior 1.0 and beyond [J]. Zebrafish, 2013, 10(1): 70-86. doi: 10.1089/zeb.2012.0861
[40] FITZGERALD J A, KÖNEMANN S, KRÜMPELMANN L, et al. Approaches to test the neurotoxicity of environmental contaminants in the zebrafish model: From behavior to molecular mechanisms [J]. Environmental Toxicology and Chemistry, 2021, 40(4): 989-1006. doi: 10.1002/etc.4951
[41] KALUEFF A V, STEWART A M, GERLAI R. Zebrafish as an emerging model for studying complex brain disorders [J]. Trends in Pharmacological Sciences, 2014, 35(2): 63-75. doi: 10.1016/j.tips.2013.12.002
[42] TEGELENBOSCH R A J, NOLDUS L P J J, RICHARDSON M K, et al. Zebrafish embryos and larvae in behavioural assays [J]. Behaviour, 2012, 149(10/11/12): 1241-1281.
[43] VORHEES C V, WILLIAMS M T, HAWKEY A B, et al. Translating neurobehavioral toxicity across species from zebrafish to rats to humans: Implications for risk assessment [J]. Frontiers in Toxicology, 2021, 3: 629229. doi: 10.3389/ftox.2021.629229
[44] JAREMA K A, HUNTER D L, SHAFFER R M, et al. Acute and developmental behavioral effects of flame retardants and related chemicals in zebrafish [J]. Neurotoxicology and Teratology, 2015, 52: 194-209. doi: 10.1016/j.ntt.2015.08.010
[45] GEIER M C, JAMES MINICK D, TRUONG L, et al. Systematic developmental neurotoxicity assessment of a representative PAH Superfund mixture using zebrafish [J]. Toxicology and Applied Pharmacology, 2018, 354: 115-125. doi: 10.1016/j.taap.2018.03.029
[46] DACH K, YAGHOOBI B, SCHMUCK M R, et al. Teratological and behavioral screening of the national toxicology program 91-compound library in zebrafish (Danio rerio) [J]. Toxicological Sciences, 2019, 167(1): 77-91. doi: 10.1093/toxsci/kfy266
[47] HAGSTROM D, TRUONG L, ZHANG S Q, et al. Comparative analysis of zebrafish and planarian model systems for developmental neurotoxicity screens using an 87-compound library [J]. Toxicological Sciences, 2019, 167(1): 15-25. doi: 10.1093/toxsci/kfy180
[48] QUEVEDO C, BEHL M, RYAN K, et al. Detection and prioritization of developmentally neurotoxic and/or neurotoxic compounds using zebrafish [J]. Toxicological Sciences, 2019, 168(1): 225-240. doi: 10.1093/toxsci/kfy291
[49] NOYES P D, HAGGARD D E, GONNERMAN G D, et al. Advanced morphological—Behavioral test platform reveals neurodevelopmental defects in embryonic zebrafish exposed to comprehensive suite of halogenated and organophosphate flame retardants [J]. Toxicological Sciences, 2015, 145(1): 177-195. doi: 10.1093/toxsci/kfv044
[50] ZHANG K, LIANG J H, BRUN N R, et al. Rapid zebrafish behavioral profiling assay accelerates the identification of environmental neurodevelopmental toxicants [J]. Environmental Science & Technology, 2021, 55(3): 1919-1929.
[51] VLIET S M, HO T C, VOLZ D C. Behavioral screening of the LOPAC1280 library in zebrafish embryos [J]. Toxicology and Applied Pharmacology, 2017, 329: 241-248. doi: 10.1016/j.taap.2017.06.011
[52] RIHEL J, PROBER D A, ARVANITES A, et al. Zebrafish behavioral profiling links drugs to biological targets and rest/wake regulation [J]. Science, 2010, 327(5963): 348-351. doi: 10.1126/science.1183090
[53] KOKEL D, BRYAN J, LAGGNER C, et al. Rapid behavior-based identification of neuroactive small molecules in the zebrafish [J]. Nature Chemical Biology, 2010, 6(3): 231-237. doi: 10.1038/nchembio.307
[54] BRUNI G, RENNEKAMP A J, VELENICH A, et al. Zebrafish behavioral profiling identifies multitarget antipsychotic-like compounds [J]. Nature Chemical Biology, 2016, 12(7): 559-566. doi: 10.1038/nchembio.2097
[55] BURGESS H A, GRANATO M. Modulation of locomotor activity in larval zebrafish during light adaptation[J]. The Journal of Experimental Biology, 2007, 210(Pt 14): 2526-2539.
[56] DALEY M C, MENDE U, CHOI B R, et al. Beyond pharmaceuticals: Fit-for-purpose new approach methodologies for environmental cardiotoxicity testing [J]. ALTEX, 2023, 40(1): 103-116.
[57] HODGSON P, IRELAND J, GRUNOW B. Fish, the better model in human heart research? Zebrafish Heart aggregates as a 3D spontaneously cardiomyogenic in vitro model system [J]. Progress in Biophysics and Molecular Biology, 2018, 138: 132-141. doi: 10.1016/j.pbiomolbio.2018.04.009
[58] DHILLON S S, DÓRÓ E, MAGYARY I, et al. Optimisation of embryonic and larval ECG measurement in zebrafish for quantifying the effect of QT prolonging drugs [J]. PLoS One, 2013, 8(4): e60552. doi: 10.1371/journal.pone.0060552
[59] LIN M H, CHOU H C, CHEN Y F, et al. Development of a rapid and economic in vivo electrocardiogram platform for cardiovascular drug assay and electrophysiology research in adult zebrafish [J]. Scientific Reports, 2018, 8(1): 1-12.
[60] LIU C C, LI L, LAM Y W, et al. Improvement of surface ECG recording in adult zebrafish reveals that the value of this model exceeds our expectation [J]. Scientific Reports, 2016, 6(1): 25073. doi: 10.1038/srep25073
[61] de LUCA E, ZACCARIA G M, HADHOUD M, et al. ZebraBeat: A flexible platform for the analysis of the cardiac rate in zebrafish embryos [J]. Scientific Reports, 2014, 4(1): 4898. doi: 10.1038/srep04898
[62] LETAMENDIA A, QUEVEDO C, IBARBIA I, et al. Development and validation of an automated high-throughput system for zebrafish in vivo screenings [J]. PLoS One, 2012, 7(5): e36690. doi: 10.1371/journal.pone.0036690
[63] BURNS C G, MILAN D J, GRANDE E J, et al. High-throughput assay for small molecules that modulate zebrafish embryonic heart rate [J]. Nature Chemical Biology, 2005, 1(5): 263-264. doi: 10.1038/nchembio732
[64] CORNET C, CALZOLARI S, MIÑANA-PRIETO R, et al. ZeGlobalTox: An innovative approach to address organ drug toxicity using zebrafish [J]. International Journal of Molecular Sciences, 2017, 18(4): 864. doi: 10.3390/ijms18040864
[65] MILAN D J, PETERSON T A, RUSKIN J N, et al. Drugs that induce repolarization abnormalities cause bradycardia in zebrafish [J]. Circulation, 2003, 107(10): 1355-1358. doi: 10.1161/01.CIR.0000061912.88753.87
[66] DYBALLA S, MIÑANA R, RUBIO-BROTONS M, et al. Comparison of zebrafish larvae and hiPSC cardiomyocytes for predicting drug-induced cardiotoxicity in humans [J]. Toxicological Sciences, 2019, 171(2): 283-295. doi: 10.1093/toxsci/kfz165
[67] MENG H Y, LIANG J H, ZHENG X H, et al. Using a high-throughput zebrafish embryo screening approach to support environmental hazard ranking for cardiovascular agents [J]. Science of the Total Environment, 2020, 702: 134703. doi: 10.1016/j.scitotenv.2019.134703
[68] XING X M, KANG J M, QIU J H, et al. Waterborne exposure to low concentrations of BDE-47 impedes early vascular development in zebrafish embryos/larvae [J]. Aquatic Toxicology, 2018, 203: 19-27. doi: 10.1016/j.aquatox.2018.07.012
[69] ZHONG X L, QIU J H, KANG J M, et al. Exposure to tris(1, 3-dichloro-2-propyl) phosphate (TDCPP) induces vascular toxicity through Nrf2-VEGF pathway in zebrafish and human umbilical vein endothelial cells [J]. Environmental Pollution, 2019, 247: 293-301. doi: 10.1016/j.envpol.2018.12.066
[70] LÖHR H, HAMMERSCHMIDT M. Zebrafish in endocrine systems: Recent advances and implications for human disease [J]. Annual Review of Physiology, 2011, 73: 183-211. doi: 10.1146/annurev-physiol-012110-142320
[71] HERZOG W, ZENG X C, LELE Z, et al. Adenohypophysis formation in the zebrafish and its dependence on sonic hedgehog [J]. Developmental Biology, 2003, 254(1): 36-49. doi: 10.1016/S0012-1606(02)00124-0
[72] MOURIEC K, LAREYRE J J, TONG S K, et al. Early regulation of brain aromatase (cyp19a1b) by estrogen receptors during zebrafish development [J]. Developmental Dynamics:an Official Publication of the American Association of Anatomists, 2009, 238(10): 2641-2651. doi: 10.1002/dvdy.22069
[73] TONG S K, MOURIEC K, KUO M W, et al. A cyp19a1b-gfp (aromatase B) transgenic zebrafish line that expresses GFP in radial glial cells [J]. Genesis, 2009, 47(2): 67-73. doi: 10.1002/dvg.20459
[74] PORAZZI P, CALEBIRO D, BENATO F, et al. Thyroid gland development and function in the zebrafish model [J]. Molecular and Cellular Endocrinology, 2009, 312(1/2): 14-23.
[75] GORELICK D A, HALPERN M E. Visualization of estrogen receptor transcriptional activation in zebrafish [J]. Endocrinology, 2011, 152(7): 2690-2703. doi: 10.1210/en.2010-1257
[76] LEE O, TAKESONO A, TADA M, et al. Biosensor zebrafish provide new insights into potential health effects of environmental estrogens [J]. Environmental Health Perspectives, 2012, 120(7): 990-996. doi: 10.1289/ehp.1104433
[77] CHEN H, HU J Y, YANG J, et al. Generation of a fluorescent transgenic zebrafish for detection of environmental estrogens [J]. Aquatic Toxicology, 2010, 96(1): 53-61. doi: 10.1016/j.aquatox.2009.09.015
[78] JI C, JIN X, HE J Y, et al. Use of TSHβ: EGFP transgenic zebrafish as a rapid in vivo model for assessing thyroid-disrupting chemicals [J]. Toxicology and Applied Pharmacology, 2012, 262(2): 149-155. doi: 10.1016/j.taap.2012.04.029
[79] BENATO F, COLLETTI E, SKOBO T, et al. A living biosensor model to dynamically trace glucocorticoid transcriptional activity during development and adult life in zebrafish [J]. Molecular and Cellular Endocrinology, 2014, 392(1/2): 60-72.
[80] GREEN J M, METZ J, LEE O, et al. High-content and semi-automated quantification of responses to estrogenic chemicals using a novel translucent transgenic zebrafish [J]. Environmental Science & Technology, 2016, 50(12): 6536-6545.
[81] VERNETTI L A, VOGT A, GOUGH A, et al. Evolution of experimental models of the liver to predict human drug hepatotoxicity and efficacy [J]. Clinics in Liver Disease, 2017, 21(1): 197-214. doi: 10.1016/j.cld.2016.08.013
[82] HILL A, MESENS N, STEEMANS M, et al. Comparisons between in vitro whole cell imaging and in vivo zebrafish-based approaches for identifying potential human hepatotoxicants earlier in pharmaceutical development [J]. Drug Metabolism Reviews, 2012, 44(1): 127-140. doi: 10.3109/03602532.2011.645578
[83] GOESSLING W, SADLER K C. Zebrafish: An important tool for liver disease research [J]. Gastroenterology, 2015, 149(6): 1361-1377. doi: 10.1053/j.gastro.2015.08.034
[84] de SOUZA ANSELMO C, SARDELA V F, de SOUSA V P, et al. Zebrafish (Danio rerio): A valuable tool for predicting the metabolism of xenobiotics in humans? [J]. Comparative Biochemistry and Physiology Part C:Toxicology & Pharmacology, 2018, 212: 34-46.
[85] POON K L, WANG X G, NG A S, et al. Humanizing the zebrafish liver shifts drug metabolic profiles and improves pharmacokinetics of CYP3A4 substrates [J]. Archives of Toxicology, 2017, 91(3): 1187-1197. doi: 10.1007/s00204-016-1789-5
[86] ALZUALDE A, BEHL M, SIPES N S, et al. Toxicity profiling of flame retardants in zebrafish embryos using a battery of assays for developmental toxicity, neurotoxicity, cardiotoxicity and hepatotoxicity toward human relevance [J]. Neurotoxicology and Teratology, 2018, 70: 40-50. doi: 10.1016/j.ntt.2018.10.002
[87] DASGUPTA S, REDDAM A, LIU Z K, et al. High-content screening in zebrafish identifies perfluorooctanesulfonamide as a potent developmental toxicant [J]. Environmental Pollution, 2020, 256: 113550. doi: 10.1016/j.envpol.2019.113550
[88] OU H C, SANTOS F, RAIBLE D W, et al. Drug screening for hearing loss: Using the zebrafish lateral line to screen for drugs that prevent and cause hearing loss [J]. Drug Discovery Today, 2010, 15(7/8): 265-271.
[89] KIM S A, KIM L, KIM T H, et al. Assessing the size-dependent effects of microplastics on zebrafish larvae through fish lateral line system and gut damage[J]. Marine Pollution Bulletin, 2022, 185(Pt A): 114279.
[90] YEN H J, HORNG J L, YU C H, et al. Toxic effects of silver and copper nanoparticles on lateral-line hair cells of zebrafish embryos [J]. Aquatic Toxicology, 2019, 215: 105273. doi: 10.1016/j.aquatox.2019.105273
[91] HERNÁNDEZ P P, MORENO V, OLIVARI F A, et al. Sub-lethal concentrations of waterborne copper are toxic to lateral line neuromasts in zebrafish (Danio rerio) [J]. Hearing Research, 2006, 213(1/2): 1-10.
[92] HAYASHI L, SHETH M, YOUNG A, et al. The effect of the aquatic contaminants bisphenol-A and PCB-95 on the zebrafish lateral line [J]. NeuroToxicology, 2015, 46: 125-136. doi: 10.1016/j.neuro.2014.12.010
[93] LIN L Y, HUNG G Y, YEH Y H, et al. Acidified water impairs the lateral line system of zebrafish embryos [J]. Aquatic Toxicology, 2019, 217: 105351. doi: 10.1016/j.aquatox.2019.105351
[94] LIN L Y, ZHENG J A, HUANG S C, et al. Ammonia exposure impairs lateral-line hair cells and mechanotransduction in zebrafish embryos [J]. Chemosphere, 2020, 257: 127170. doi: 10.1016/j.chemosphere.2020.127170
[95] YOUNG A, KOCHENKOV V, McINTYRE J K, et al. Urban stormwater runoff negatively impacts lateral line development in larval zebrafish and salmon embryos [J]. Scientific Reports, 2018, 8(1): 2830. doi: 10.1038/s41598-018-21209-z
[96] ADATTO I, LAWRENCE C, THOMPSON M, et al. A new system for the rapid collection of large numbers of developmentally staged zebrafish embryos [J]. PLoS One, 2011, 6(6): e21715. doi: 10.1371/journal.pone.0021715
[97] CARVALHO R, de SONNEVILLE J, STOCKHAMMER O W, et al. A high-throughput screen for tuberculosis progression [J]. PLoS One, 2011, 6(2): e16779. doi: 10.1371/journal.pone.0016779
[98] WANG G L, RAJPUROHIT S K, DELASPRE F, et al. First quantitative high-throughput screen in zebrafish identifies novel pathways for increasing pancreatic β-cell mass [J]. eLife, 2015, 4: e08261. doi: 10.7554/eLife.08261
[99] ZHANG X P, LU Z, GELINAS D, et al. Batch transfer of zebrafish embryos into multiwell plates [J]. IEEE Transactions on Automation Science and Engineering, 2011, 8(3): 625-632. doi: 10.1109/TASE.2011.2121903
[100] FUAD N M, SKOMMER J, FRIEDRICH T, et al. An integrated micromechanical large particle in flow sorter (MILPIS)[C]//SPIE Proceedings, Bio-MEMS and Medical Microdevices II. Barcelona, Spain. SPIE, 2015.
[101] KAUFMAN C K, WHITE R M, ZON L. Chemical genetic screening in the zebrafish embryo [J]. Nature Protocols, 2009, 4(10): 1422-1432. doi: 10.1038/nprot.2009.144
[102] ZHU F, SKOMMER J, HUANG Y S, et al. Fishing on chips: Up-and-coming technological advances in analysis of zebrafish and Xenopus embryos [J]. Cytometry. Part A:the Journal of the International Society for Analytical Cytology, 2014, 85(11): 921-932. doi: 10.1002/cyto.a.22571
[103] YANG F, GAO C, WANG P, et al. Fish-on-a-chip: Microfluidics for zebrafish research [J]. Lab on a Chip, 2016, 16(7): 1106-1125. doi: 10.1039/C6LC00044D
[104] AKAGI J, KHOSHMANESH K, EVANS B, et al. Miniaturized embryo array for automated trapping, immobilization and microperfusion of zebrafish embryos [J]. PLoS One, 2012, 7(5): e36630. doi: 10.1371/journal.pone.0036630
[105] ZHU F, WIGH A, FRIEDRICH T, et al. Automated lab-on-a-chip technology for fish embryo toxicity tests performed under continuous microperfusion (μFET) [J]. Environmental Science & Technology, 2015, 49(24): 14570-14578.
[106] LI Y B, YANG X J, CHEN Z G, et al. Comparative toxicity of lead (Pb2+), copper (Cu2+), and mixtures of lead and copper to zebrafish embryos on a microfluidic chip [J]. Biomicrofluidics, 2015, 9(2): 024105. doi: 10.1063/1.4913699
[107] LI Y B, YANG F, CHEN Z G, et al. Zebrafish on a chip: A novel platform for real-time monitoring of drug-induced developmental toxicity [J]. PLoS One, 2014, 9(4): e94792. doi: 10.1371/journal.pone.0094792
[108] CHOUDHURY D, van NOORT D, ILIESCU C, et al. Fish and Chips: A microfluidic perfusion platform for monitoring zebrafish development [J]. Lab on a Chip, 2012, 12(5): 892-900. doi: 10.1039/C1LC20351G
[109] YANG F, CHEN Z G, PAN J B, et al. An integrated microfluidic array system for evaluating toxicity and teratogenicity of drugs on embryonic zebrafish developmental dynamics [J]. Biomicrofluidics, 2011, 5(2): 24115. doi: 10.1063/1.3605509
[110] WANG K I K, SALCIC Z, YEH J, et al. Toward embedded laboratory automation for smart lab-on-a-chip embryo arrays [J]. Biosensors and Bioelectronics, 2013, 48: 188-196. doi: 10.1016/j.bios.2013.04.033
[111] ZHAO Y L, SUN H, SHA X P, et al. A review of automated microinjection of zebrafish embryos [J]. Micromachines, 2018, 10(1): 7. doi: 10.3390/mi10010007
[112] ZHU F, HALL C J, CROSIER P S, et al. Interfacing lab-on-a-chip embryo technology with high-definition imaging cytometry [J]. Zebrafish, 2015, 12(4): 315-318. doi: 10.1089/zeb.2015.1105
[113] MARTINEAU P R, MOURRAIN P. Tracking zebrafish larvae in group–Status and perspectives [J]. Methods, 2013, 62(3): 292-303. doi: 10.1016/j.ymeth.2013.05.002
[114] ZHOU Y Z, CATTLEY R T, CARIO C L, et al. Quantification of larval zebrafish motor function in multiwell plates using open-source MATLAB applications [J]. Nature Protocols, 2014, 9(7): 1533-1548. doi: 10.1038/nprot.2014.094
[115] INGEBRETSON J J, MASINO M A. Quantification of locomotor activity in larval zebrafish: Considerations for the design of high-throughput behavioral studies [J]. Frontiers in Neural Circuits, 2013, 7: 109.
[116] FONTAINE E, LENTINK D, KRANENBARG S, et al. Automated visual tracking for studying the ontogeny of zebrafish swimming[J]. The Journal of Experimental Biology, 2008, 211(Pt 8): 1305-1316.
[117] PARDO-MARTIN C, CHANG T Y, KOO B K, et al. High-throughput in vivo vertebrate screening [J]. Nature Methods, 2010, 7(8): 634-636. doi: 10.1038/nmeth.1481
[118] CHANG T Y, PARDO-MARTIN C, ALLALOU A, et al. Fully automated cellular-resolution vertebrate screening platform with parallel animal processing [J]. Lab on a Chip, 2012, 12(4): 711-716. doi: 10.1039/C1LC20849G
[119] PULAK R. Tools for automating the imaging of zebrafish larvae [J]. Methods, 2016, 96: 118-126. doi: 10.1016/j.ymeth.2015.11.021
[120] EARLY J J, COLE K L, WILLIAMSON J M, et al. An automated high-resolution in vivo screen in zebrafish to identify chemical regulators of myelination [J]. eLife, 2018, 7: e35136. doi: 10.7554/eLife.35136
[121] CHEN C Y, GU Y Y, PHILIPPE J, et al. Acoustofluidic rotational tweezing enables high-speed contactless morphological phenotyping of zebrafish larvae [J]. Nature Communications, 2021, 12(1): 1-13. doi: 10.1038/s41467-020-20314-w
[122] YANG G, WANG L B, QIN X F, et al. Heterogeneities of zebrafish vasculature development studied by a high throughput light-sheet flow imaging system [J]. Biomedical Optics Express, 2022, 13(10): 5344-5357. doi: 10.1364/BOE.470058
[123] KELLER P J, SCHMIDT A D, WITTBRODT J, et al. Reconstruction of zebrafish early embryonic development by scanned light sheet microscopy [J]. Science, 2008, 322(5904): 1065-1069. doi: 10.1126/science.1162493
[124] ALLALOU A, WU Y L, GHANNAD-REZAIE M, et al. Automated deep-phenotyping of the vertebrate brain [J]. eLife, 2017, 6: e23379. doi: 10.7554/eLife.23379
[125] PARDO-MARTIN C, ALLALOU A, MEDINA J, et al. High-throughput hyperdimensional vertebrate phenotyping [J]. Nature Communications, 2013, 4(1): 1-9.
[126] BOUTROS M, HEIGWER F, LAUFER C. Microscopy-based high-content screening [J]. Cell, 2015, 163(6): 1314-1325. doi: 10.1016/j.cell.2015.11.007
[127] MIKUT R, DICKMEIS T, DRIEVER W, et al. Automated processing of zebrafish imaging data: A survey [J]. Zebrafish, 2013, 10(3): 401-421. doi: 10.1089/zeb.2013.0886
[128] OTTERSTROM J J, LUBIN A, PAYNE E M, et al. Technologies bringing young Zebrafish from a niche field to the limelight [J]. SLAS Technology, 2022, 27(2): 109-120. doi: 10.1016/j.slast.2021.12.005
[129] LUBIN A, OTTERSTROM J, HOADE Y, et al. A versatile, automated and high-throughput drug screening platform for zebrafish embryos [J]. Biology Open, 2021, 10(9): bio058513. doi: 10.1242/bio.058513
[130] WESTHOFF J H, GISELBRECHT S, SCHMIDTS M, et al. Development of an automated imaging pipeline for the analysis of the zebrafish larval kidney [J]. PLoS One, 2013, 8(12): e82137. doi: 10.1371/journal.pone.0082137
[131] AKERBERG A A, BURNS C E, BURNS C G, et al. Deep learning enables automated volumetric assessments of cardiac function in zebrafish [J]. Disease Models & Mechanisms, 2019, 12(10): dmm040188.
[132] RONNEBERGER O, LIU K, RATH M, et al. ViBE-Z: A framework for 3D virtual colocalization analysis in zebrafish larval brains [J]. Nature Methods, 2012, 9(7): 735-742. doi: 10.1038/nmeth.2076