CRISPR-Cas9, CRISPRi en zebravissen. Een zee aan informatie

Pieter
Maene

CRISPR-Cas is een term die voor velen al een belletje doet rinkelen. Dit komt doordat CRISPR-Cas

gebaseerde methoden niet meer weg te denken zijn uit moderne laboratoria. Het vindt

toepassingen in landbouw, biotechnologie, geneeskunde en farmaceutica.

Het CRISPR-Cas systeem is onderdeel van wat we “gerichte nucleasen” heten, enzymen die in DNA

geprogrammeerd kunnen ‘knippen’. Deze enzymen zijn voor onderzoekers en wetenschappers van

zeer groot belang en worden daarom ook vaak gebruikt. Het CRISPR-systeem blijft aanpassingen en

verbeteringen ondergaan, zo is er ondertussen een aanpassing gekomen op het Cas proteïne

waardoor deze niet meer knipt, maar wel nog gentranscriptie kan tegenhouden. Dit staat bekend als

‘gene silencing’ en de techniek wordt CRISPRi (CRISPR interference) genoemd. In een veld dat zo

sterk uitbreidt als de genetische manipulatie is het van belang om een duidelijk overzicht te houden

van de verschillende mogelijkheden.

Zebravissen zijn een modelorganisme die al jaren gebruikt worden in biomedisch onderzoek. Ze

werden voornamelijk ingezet in onderzoek naar ontwikkeling en groei. Ondertussen is aangetoond

dat de zebravissen een goed modelorganisme kunnen zijn voor immunologisch onderzoek omdat er

grote gelijkenissen zijn tussen het immuunsysteem van zebravissen en van de mens. Het gebruik van

zebravissen is in opmars door hun vele toepassingen en voordelen.

Veel biomedisch onderzoek maakt gebruik van genetische manipulatie en genetische screens om

genen te identificeren of uit te schakelen. De combinatie van CRISPR en zebravissen leidt dus tot een

zeer bruikbaar systeem om ziektemodellen op te maken. Het is aangetoond dat het CRISPR-systeem

toepasbaar is in zebravissen. Dit is dus een veelbelovend veld.

Deze theoretische bachelor proef onderzoekt ten eerste de gerichte nucleasen (ZFN, TALEN) met de

focus gericht op de CRISPR-systemen. De voor- en nadelen van de nucleasen alsook het ontstaan,

gebruik en de werkingsmechanismen. Vervolgens wordt het de zebravis (Danio rerio) als

modelorganisme onder de loep genomen. Er wordt besproken welke voordelen dit modelorganisme

heeft, maar ook welke tekortkomingen er zijn. Ten slotte wordt besproken hoe CRISPR en CRISPRi

gebruikt kunnen worden in de zebravis. Hierbij wordt de toediening besproken en wordt er ook

aandacht besteed aan de verschillende beschikbare methoden die het mogelijk maken om de

efficiëntie van de CRISPR-modificatie te controleren.

 

Bibliografie

ADDIN Mendeley Bibliography
CSL_BIBLIOGRAPHY Ablain, J., Durand, E. M., Yang, S., Zhou, Y., & Zon, L. I. (2015). A CRISPR/Cas9 Vector System for Tissue-Specific Gene Disruption in Zebrafish. Developmental Cell, 32(6), 756–764. https://doi.org/10.1016/j.devcel.2015.01.032

Adli, M. (2018). The CRISPR tool kit for genome editing and beyond. Nature Communications, 9(1). https://doi.org/10.1038/s41467-018-04252-2

Agrawal, N., Dasaradhi, P., Mohammed, A., Malhotra, P., Bhatnagar, R. K., & Mukherjee, S. K. (2003). RNA Interference: Biology, Mechanism, and Applications. Microbiology and Molecular Biology Reviews, 67(4), 657–685. https://doi.org/10.1128/MMBR.67.4.657

Albadri, S., De Santis, F., Di Donato, V., & Del Bene, F. (2017). CRISPR/Cas9-Mediated Knockin and Knockout in Zebrafish. In Research and Perspectives in Neurosciences (Issue 9783319601915, pp. 41–49). Springer Verlag. https://doi.org/10.1007/978-3-319-60192-2_4

Allen, J. P., & Neely, M. N. (2010). Trolling for the ideal model host: zebrafish take the bait. Future Microbiology, 5(4), 563–569. https://doi.org/10.2217/fmb.10.24

Alvarado Bremer, J. R., Smith, B. L., Moulton, D. L., Lu, C. P., & Cornic, M. (2014). Shake and stew: A non-destructive PCR-ready DNA isolation method from a single preserved fish larva. Journal of Fish Biology, 84(1), 267–272. https://doi.org/10.1111/jfb.12280

Amatruda, J. F., Shepard, J. L., Stern, H. M., & Zon, L. I. (2002). Zebrafish as a cancer model system. Cancer Cell, 1(3), 229–231. https://doi.org/10.1016/S1535-6108(02)00052-1

Amitai, G., & Sorek, R. (2016). CRISPR-Cas adaptation: Insights into the mechanism of action. Nature Reviews Microbiology, 14(2), 67–76. https://doi.org/10.1038/nrmicro.2015.14

Amsterdam, A., Burgess, S., Golling, G., Chen, W., Sun, Z., Townsend, K., Farrington, S., Haldi, M., & Hopkins, N. (1999). A large-scale insertional mutagenesis screen in zebrafish. Genes and Development, 13(20), 2713–2724. https://doi.org/10.1101/gad.13.20.2713

Aranishi, F. (2006). Single fish egg DNA extraction for PCR amplification. Conservation Genetics, 7(1), 153–156. https://doi.org/10.1007/s10592-005-5387-y

Auer, T. O., Duroure, K., De Cian, A., Concordet, J.-P., & Del Bene, F. (2014). Highly efficient CRISPR/Cas9-mediated knock-in in zebrafish by homology-independent DNA repair. Genome Research, 24(1), 142–153. https://doi.org/10.1101/gr.161638.113

Babon, J. J., McKenzie, M., & Cotton, R. G. H. (2003). The Use of Resolvases T4 Endonuclease VII and T7 Endonuclease I in Mutation Detection. Molecular Biotechnology, 23(1), 73–82. https://doi.org/10.1385/MB:23:1:73

Bambino, K., & Chu, J. (2017). Zebrafish in Toxicology and Environmental Health. In Current Topics in Developmental Biology (Vol. 124, pp. 331–367). Academic Press Inc. https://doi.org/10.1016/bs.ctdb.2016.10.007

Bannikov, A. V., & Lavrov, A. V. (2017). CRISPR/CAS9, the king of genome editing tools. Molecular Biology, 51(4), 514–525. https://doi.org/10.1134/S0026893317040033

Baum, C., Kustikova, O., Modlich, U., Li, Z., & Fehse, B. (2006). Mutagenesis and oncogenesis by chromosomal insertion of gene transfer vectors. Human Gene Therapy, 17(3), 253–263. https://doi.org/10.1089/hum.2006.17.253

Baum, C., von Kalle, C., Staal, F. J. T., Li, Z., Fehse, B., Schmidt, M., Weerkamp, F., Karlsson, S., Wagemaker, G., & Williams, D. A. (2004). Chance or necessity? Insertional Mutagenesis in Gene Therapy and Its Consequences. Molecular Therapy, 9(1), 5–13. https://doi.org/10.1016/j.ymthe.2003.10.013

Bertrand, J. Y., & Traver, D. (2009). Hematopoietic cell development in the zebrafish embryo. Current Opinion in Hematology, 16(4), 243–248. https://doi.org/10.1097/MOH.0b013e32832c05e4

Beumer, K. J., Trautman, J. K., Bozas, A., Liu, J. L., Rutter, J., Gall, J. G., & Carroll, D. (2008). Efficient gene targeting in Drosophila by direct embryo injection with zinc-finger nucleases. Proceedings of the National Academy of Sciences of the United States of America, 105(50), 19821–19826. https://doi.org/10.1073/pnas.0810475105

Bi, H., & Yang, B. (2017). Gene Editing With TALEN and CRISPR/Cas in Rice. In Progress in Molecular Biology and Translational Science (1st ed., Vol. 149, pp. 81–98). Elsevier Inc. https://doi.org/10.1016/bs.pmbts.2017.04.006

Bitinaite, J., Wah, D. A., Aggarwal, A. K., & Schildkraut, I. (1998). Foki dimerization is required for dna cleavage. Proceedings of the National Academy of Sciences of the United States of America, 95(18), 10570–10575. https://doi.org/10.1073/pnas.95.18.10570

Boch, J., Scholze, H., Schornack, S., Landgraf, A., Hahn, S., Kay, S., Lahaye, T., Nickstadt, A., & Bonas, U. (2009). Breaking the code of DNA binding specificity of TAL-type III effectors. Science, 326(5959), 1509–1512. https://doi.org/10.1126/science.1178811

Bogdanove, A. J., & Voytas, D. F. (2011). TAL effectors: Customizable proteins for DNA targeting. Science, 333(6051), 1843–1846. https://doi.org/10.1126/science.1204094

Bonas, U. (1990). Isolation of a gene cluster from Xanthomonas campestris pv. vesicatoria that determines pathogenicity and the hypersensitive response on pepper and tomato.pdf. Mol Plant Microbe Interact, 4, 81–88. https://doi.org/10005382285

Bouard, D., Alazard-Dany, N., & Cosset, F.-L. (2009). Viral vectors: from virology to transgene expression. British Journal of Pharmacology, 157(2), 153–165. https://doi.org/10.1038/bjp.2008.349

Broughton, J. P., Deng, X., Yu, G., Fasching, C. L., Servellita, V., Singh, J., Miao, X., Streithorst, J. A., Granados, A., Sotomayor-gonzalez, A., Zorn, K., Gopez, A., Hsu, E., Gu, W., Miller, S., Pan, C., Guevara, H., Wadford, D. A., Chen, J. S., & Chiu, C. Y. (2019). CRISPR – Cas12-based detection of SARS-CoV-2.

Burger, A., Lindsay, H., Felker, A., Hess, C., Anders, C., Chiavacci, E., Zaugg, J., Weber, L. M., Catena, R., Jinek, M., Robinson, M. D., & Mosimann, C. (2016). Maximizing mutagenesis with solubilized CRISPR-Cas9 ribonucleoprotein complexes. Development, 143(11), 2025–2037. https://doi.org/10.1242/dev.134809

Carroll, D. (2011). Genome Engineering With Zinc-Finger Nucleases. https://doi.org/10.1534/genetics.111.131433

Chandrasegaran, S., & Carroll, D. (2016). Origins of Programmable Nucleases for Genome Engineering. Journal of Molecular Biology, 428(5), 963–989. https://doi.org/10.1016/j.jmb.2015.10.014

Chandrasekaran, A. P., Song, M., Kim, K. S., & Ramakrishna, S. (2018). Different Methods of Delivering CRISPR/Cas9 Into Cells. In Progress in Molecular Biology and Translational Science (1st ed., Vol. 159). Elsevier Inc. https://doi.org/10.1016/bs.pmbts.2018.05.001

Chang, H. H. Y., Pannunzio, N. R., Adachi, N., & Lieber, M. R. (2017). Non-homologous DNA end joining and alternative pathways to double-strand break repair HHS Public Access. Nat Rev Mol Cell Biol, 18(8), 495–506. https://doi.org/10.1038/nrm.2017.48

Chang, N., Sun, C., Gao, L., Zhu, D., Xu, X., Zhu, X., Xiong, J. W., & Xi, J. J. (2013). Genome editing with RNA-guided Cas9 nuclease in Zebrafish embryos. Cell Research, 23(4), 465–472. https://doi.org/10.1038/cr.2013.45

Chen, J., Zhang, X., Wang, T., Li, Z., Guan, G., & Hong, Y. (2012). Efficient detection, quantification and enrichment of subtle allelic alterations. DNA Research, 19(5), 423–433. https://doi.org/10.1093/dnares/dss023

Chowdhury, T. A., Koceja, C., Eisa-Beygi, S., Kleinstiver, B. P., Kumar, S. N., Lin, C. W., Li, K., Prabhudesai, S., Joung, J. K., & Ramchandran, R. (2018). Temporal and spatial post-transcriptional regulation of zebrafish tie1 mRNA by Long Noncoding RNA during brain vascular assembly. Arteriosclerosis, Thrombosis, and Vascular Biology, 38(7), 1562–1575. https://doi.org/10.1161/ATVBAHA.118.310848

Chowdhury, T. A., Li, K., Ramchandran, R., Daniel, J. F. N., David L, K. S., David H, W., Holmes, Chowdhury, T. A., Li, K., Ramchandran, R., Daniel, J. F. N., David L, K. S., David H, W., & Holmes. (2019). Lessons learned from a lncRNA odyssey for two genes with vascular functions, DLL4 and TIE1. Vascular Pharmacology, 114(1), 103–109. https://doi.org/10.1016/j.vph.2018.06.010

Christian, M., Cermak, T., Doyle, E. L., Schmidt, C., Zhang, F., Hummel, A., Bogdanove, A. J., & Voytas, D. F. (2010). Targeting DNA Double-Strand Breaks with TAL Effector Nucleases. Genetics, 186(2), 757–761. https://doi.org/10.1534/genetics.110.120717

Chylinski, K., Makarova, K. S., Charpentier, E., & Koonin, E. V. (2014). Classification and evolution of type II CRISPR-Cas systems. Nucleic Acids Research, 42(10), 6091–6105. https://doi.org/10.1093/nar/gku241

Condreay, J. P., Witherspoon, S. M., Clay, W. C., & Kost, T. A. (1999). Transient and stable gene expression in mammalian cells transduced with a recombinant baculovirus vector. Proceedings of the National Academy of Sciences of the United States of America, 96(1), 127–132. https://doi.org/10.1073/pnas.96.1.127

Cong, L., Ran, F. A., Cox, D., Lin, S., Barretto, R., Habib, N., Hsu, P. D., Wu, X., Jiang, W., Marraffini, L. A., & Zhang, F. (2013). Multiplex Genome Engineering Using CRISPR/Cas Systems. Science, 339(6121), 819–823. https://doi.org/10.1126/science.1231143

Davidson, A. J., & Zon, L. I. (2004). The “definitive” (and ’primitive’) guide to zebrafish hematopoiesis. In Oncogene (Vol. 23, Issue 43 REV. ISS. 6, pp. 7233–7246). Nature Publishing Group. https://doi.org/10.1038/sj.onc.1207943

Dobson, J. (2006). Gene therapy progress and prospects: Magnetic nanoparticle-based gene delivery. In Gene Therapy (Vol. 13, Issue 4, pp. 283–287). Nature Publishing Group. https://doi.org/10.1038/sj.gt.3302720

Dong, J., Fan, P., & Frizzell, R. A. (1996). Quantitative Analysis of the Packaging Capacity of Recombinant Adeno-Associated Virus. Human Gene Therapy, 7(17), 2101–2112. https://doi.org/10.1089/hum.1996.7.17-2101

Doudna, J. A., & Charpentier, E. (2014). The new frontier of genome engineering with CRISPR-Cas9. Science, 346(6213). https://doi.org/10.1126/science.1258096

Doyon, Y., McCammon, J. M., Miller, J. C., Faraji, F., Ngo, C., Katibah, G. E., Amora, R., Hocking, T. D., Zhang, L., Rebar, E. J., Gregory, P. D., Urnov, F. D., & Amacher, S. L. (2008). Heritable targeted gene disruption in zebrafish using designed zinc-finger nucleases. Nature Biotechnology, 26(6), 702–708. https://doi.org/10.1038/nbt1409

Driever, W., Solnica-Krezel, L., Schier, A. F., Neuhauss, S. C. F., Malicki, J., Stemple, D. L., Stainier, D. Y. R., Zwartkruis, F., Abdelilah, S., Rangini, Z., Belak, J., & Boggs, C. (1996). A genetic screen for mutations affecting embryogenesis in zebrafish. Development, 123, 37–46. https://doi.org/10.5167/uzh-215

Eisen, J. S., & Smith, J. C. (2008). Controlling morpholino experiments: don’t stop making antisense. Development, 135(10), 1735–1743. https://doi.org/10.1242/dev.001115

El-Brolosy, M. A., & Stainier, D. Y. R. (2017). Genetic compensation: A phenomenon in search of mechanisms. PLOS Genetics, 13(7), e1006780. https://doi.org/10.1371/journal.pgen.1006780

Engeszer, R. E., Patterson, L. B., Rao, A. A., & Parichy, D. M. (2007). Zebrafish in The Wild: A Review of Natural History And New Notes from The Field. Zebrafish, 4(1), 21–40. https://doi.org/10.1089/zeb.2006.9997

Fineran, P. C., & Charpentier, E. (2012). Memory of viral infections by CRISPR-Cas adaptive immune systems: Acquisition of new information. Virology, 434(2), 202–209. https://doi.org/10.1016/j.virol.2012.10.003

Fire, A., Xu, S., Montgomery, M. K., Kostas, S. A., Driver, S. E., & Mello, C. C. (1998). Potent andspecific genetic interferenceby double-strandedRNA in Caenorhabditiselegans. Nature, 391(February), 806–811.

Foley, J. E., Yeh, J.-R. J., Maeder, M. L., Reyon, D., Sander, J. D., Peterson, R. T., & Joung, J. K. (2009). Rapid Mutation of Endogenous Zebrafish Genes Using Zinc Finger Nucleases Made by Oligomerized Pool ENgineering (OPEN). PLoS ONE, 4(2), e4348. https://doi.org/10.1371/journal.pone.0004348

Franklin, R. E., & Gosling, R. G. (1995). Molecular structure of nucleic acids. Molecular configuration in sodium thymonucleate. 1953.Franklin, R. E., & Gosling, R. G. (1995). Molecular structure of nucleic acids. Molecular configuration in sodium thymonucleate. 1953. Annals of the New York Acade. Annals of the New York Academy of Sciences, 758, 16–17. https://doi.org/10.1038/171740a0

Gagnon, J. A., Valen, E., Thyme, S. B., Huang, P., Ahkmetova, L., Pauli, A., Montague, T. G., Zimmerman, S., Richter, C., & Schier, A. F. (2014). Efficient mutagenesis by Cas9 protein-mediated oligonucleotide insertion and large-scale assessment of single-guide RNAs. PLoS ONE, 9(5), 5–12. https://doi.org/10.1371/journal.pone.0098186

Gasiunas, G., Barrangou, R., Horvath, P., & Siksnys, V. (2012). Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proceedings of the National Academy of Sciences of the United States of America, 109(39), E2579–E2586. https://doi.org/10.1073/pnas.1208507109

Gilbert, L. A., Larson, M. H., Morsut, L., Liu, Z., Brar, G. A., Torres, S. E., Stern-Ginossar, N., Brandman, O., Whitehead, E. H., Doudna, J. A., Lim, W. A., Weissman, J. S., & Qi, L. S. (2013). CRISPR-Mediated Modular RNA-Guided Regulation of Transcription in Eukaryotes. Cell. https://doi.org/10.1016/j.cell.2013.06.044

Goldsmith, J. R., & Jobin, C. (2012). Think Small: Zebrafish as a Model System of Human Pathology. Journal of Biomedicine and Biotechnology, 2012, 1–12. https://doi.org/10.1155/2012/817341

Guo, S., & Kemphues, K. J. (1995). par-1, a gene required for establishing polarity in C. elegans embryos, encodes a putative Ser/Thr kinase that is asymmetrically distributed. Cell, 81(4), 611–620. https://doi.org/10.1016/0092-8674(95)90082-9

Gupta, S. K., & Shukla, P. (2017). Gene editing for cell engineering: trends and applications. Critical Reviews in Biotechnology, 37(5), 672–684. https://doi.org/10.1080/07388551.2016.1214557

H. Meijer, A., & P. Spaink, H. (2011). Host-Pathogen Interactions Made Transparent with the Zebrafish Model. Current Drug Targets, 12(7), 1000–1017. https://doi.org/10.2174/138945011795677809

H Larson, M., A Gilbert, L., Xiaowo, W., A Lim, W., S Weissman, J., & S Qi, L. (2012). CRISPR interference (CRISPRi) for sequence-specific control of gene expression. Bone, 23(1), 1–7. https://doi.org/10.1038/jid.2014.371

Hall, C. J., Flores, M. V., Crosier, K. E., & Crosier, P. S. (2008). Live imaging early immune cell ontogeny and function in zebrafish Danio rerio. Journal of Fish Biology, 73(8), 1833–1871. https://doi.org/10.1111/j.1095-8649.2008.01980.x

Hansson, G. K., & Edfeldt, K. (2005). Toll to be paid at the gateway to the vessel wall. Arteriosclerosis, Thrombosis, and Vascular Biology, 25(6), 1085–1087. https://doi.org/10.1161/01.ATV.0000168894.43759.47

He, Y., Wang, M., Liu, M., Huang, L., Liu, C., Zhang, X., Yi, H., Cheng, A., Zhu, D., Yang, Q., Wu, Y., Zhao, X., Chen, S., Jia, R., Zhang, S., Liu, Y., Yu, Y., & Zhang, L. (2018). Cas1 and Cas2 from the type II-C CRISPR-cas system of Riemerella anatipestifer are required for spacer acquisition. Frontiers in Cellular and Infection Microbiology, 8(JUN). https://doi.org/10.3389/fcimb.2018.00195

Heasman, J., Kofron, M., & Wylie, C. (2000). β-catenin signaling activity dissected in the early Xenopus embryo: A novel antisense approach. Developmental Biology, 222(1), 124–134. https://doi.org/10.1006/dbio.2000.9720

Hilton, I. B., D’Ippolito, A. M., Vockley, C. M., Thakore, P. I., Crawford, G. E., Reddy, T. E., & Gersbach, C. A. (2015). Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nature Biotechnology, 33(5), 510–517. https://doi.org/10.1038/nbt.3199

Hockemeyer, D., Wang, H., Kiani, S., Lai, C. S., Gao, Q., Cassady, J. P., Cost, G. J., Zhang, L., Santiago, Y., Miller, J. C., Zeitler, B., Cherone, J. M., Meng, X., Hinkley, S. J., Rebar, E. J., Gregory, P. D., Urnov, F. D., & Jaenisch, R. (2011). Genetic engineering of human pluripotent cells using TALE nucleases. Nature Biotechnology, 29(8), 731–734. https://doi.org/10.1038/nbt.1927

Howe, K., Clark, M. D., Torroja, C. F., Torrance, J., Berthelot, C., Muffato, M., Collins, J. E., Humphray, S., McLaren, K., Matthews, L., McLaren, S., Sealy, I., Caccamo, M., Churcher, C., Scott, C., Barrett, J. C., Koch, R., Rauch, G.-J., White, S., … Stemple, D. L. (2013). The zebrafish reference genome sequence and its relationship to the human genome. Nature, 496(7446), 498–503. https://doi.org/10.1038/nature12111

Hruscha, A., Krawitz, P., Rechenberg, A., Heinrich, V., Hecht, J., Haass, C., & Schmid, B. (2013). Efficient CRISPR/Cas9 genome editing with low off-target effects in zebrafish. Development (Cambridge), 140(24), 4982–4987. https://doi.org/10.1242/dev.099085

Hustedt, N., & Durocher, D. (2017). The control of DNA repair by the cell cycle. Nature Cell Biology, 19(1), 1–9. https://doi.org/10.1038/ncb3452

Hwang, W. Y., Fu, Y., Reyon, D., Maeder, M. L., Shengdar, Q., Sander, J. D., Peterson, R. T., Yeh, J. J., & Keith, J. (2013). Efficient In Vivo Genome Editing Using RNA-Guided Nucleases. Nature Biotechnology, 31(3), 227–229. https://doi.org/10.1038/nbt.2501.Efficient

Hyde, J. R., Lynn, E., Humphreys, R., Musyl, M., West, A. P., & Vetter, R. (2005). Shipboard identification of fish eggs and larvae by multiplex PCR, and description of fertilized eggs of blue marlin, shortbill spearfish, and wahoo. Marine Ecology Progress Series, 286, 269–277. https://doi.org/10.3354/meps286269

Invitrogen. (n.d.). mMESSAGE mMACHINETM SP6 Transcription Kit. Retrieved May 17, 2020, from https://www.thermofisher.com/order/catalog/product/AM1340#/AM1340

Ishino, Y., Shinagawa, H., Makino, K., Amemura, M., & Nakatura, A. (1987). Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isoenzyme conversion in Escherichia coli, and identification of the gene product. Journal of Bacteriology, 169(12), 5429–5433. https://doi.org/10.1128/jb.169.12.5429-5433.1987

Janelle Espinoza, G., Michael Poland, J., & Alvarado Bremer, J. R. (2017). Genotyping live fish larvae: Non-lethal and noninvasive DNA isolation from 3-5 day old hatchlings. BioTechniques, 63(4), 181–186. https://doi.org/10.2144/000114598

Jao, L. E., Wente, S. R., & Chen, W. (2013). Efficient multiplex biallelic zebrafish genome editing using a CRISPR nuclease system. Proceedings of the National Academy of Sciences of the United States of America, 110(34), 13904–13909. https://doi.org/10.1073/pnas.1308335110

Jiang, F., & Doudna, J. A. (2017). CRISPR – Cas9 Structures and Mechanisms. 505–529.

Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., & Doudna, J. A. (2012). A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity. 91(June), 1052–1055. https://doi.org/10.1002/ajh.24462

Jinek, M., East, A., Cheng, A., Lin, S., Ma, E., & Doudna, J. (2013). RNA-programmed genome editing in human cells. ELife, 2013(2), 1–9. https://doi.org/10.7554/eLife.00471

Kampmann, M. (2018). CRISPRi and CRISPRa Screens in Mammalian Cells for Precision Biology and Medicine. ACS Chemical Biology, 13(2), 406–416. https://doi.org/10.1021/acschembio.7b00657

Kanther, M., & Rawls, J. F. (2010). Host–microbe interactions in the developing zebrafish. Current Opinion in Immunology, 22(1), 10–19. https://doi.org/10.1016/j.coi.2010.01.006

Kay, S., Hahn, S., Marois, E., Hause, G., & Bonas, U. (2007). A Bacterial Effector Acts as a Plant Transcription Factor and Induces a Cell Size Regulator. Science, 318(5850), 648–651. https://doi.org/10.1126/science.1144956

Kellner, M. J., Koob, J. G., Gootenberg, J. S., Abudayyeh, O. O., & Zhang, F. (2019). SHERLOCK: nucleic acid detection with CRISPR nucleases. Nature Protocols, 14(10), 2986–3012. https://doi.org/10.1038/s41596-019-0210-2

Kelly, T. J., & Smith, H. O. (1970). A restriction enzyme from Hemophilus influenzae II: Base sequence of the Recognition site. Journal of Molecular Biology, 51(2), 393–409. https://doi.org/10.1016/0022-2836(70)90150-6

Kim, E., Koo, T., Park, S. W., Kim, D., Kim, K., Cho, H. Y., Song, D. W., Lee, K. J., Jung, M. H., Kim, S., Kim, J. H., Kim, J. H., & Kim, J. S. (2017). In vivo genome editing with a small Cas9 orthologue derived from Campylobacter jejuni. Nature Communications, 8. https://doi.org/10.1038/ncomms14500

Kim, S., Kim, D., Cho, S. W., Kim, J., & Kim, J. S. (2014). Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Research, 24(6), 1012–1019. https://doi.org/10.1101/gr.171322.113

Kim, T. K., & Eberwine, J. H. (2010). Mammalian cell transfection: The present and the future. Analytical and Bioanalytical Chemistry, 397(8), 3173–3178. https://doi.org/10.1007/s00216-010-3821-6

Kim, Y. G., Cha, J., & Chandrasegaran, S. (1996). Hybrid restriction enzymes: Zinc finger fusions to Fok I cleavage domain. Proceedings of the National Academy of Sciences of the United States of America, 93(3), 1156–1160. https://doi.org/10.1073/pnas.93.3.1156

Kimmel, C. B., Ballard, W. W., Kimmel, S. R., Ullmann, B., & Schilling, T. F. (1995). Stages of embryonic development of the zebrafish. Developmental Dynamics, 203(3), 253–310. https://doi.org/10.1002/aja.1002030302

Kitano, H. (2004). Biological robustness. Nature Reviews Genetics, 5(11), 826–837. https://doi.org/10.1038/nrg1471

Kleinstiver, B. P., Pattanayak, V., Prew, M. S., Tsai, S. Q., Nguyen, N. T., Zheng, Z., & Joung, J. K. (2016). High-fidelity CRISPR–Cas9 nucleases with no detectable genome-wide off-target effects. Nature, 529(7587), 490–495. https://doi.org/10.1038/nature16526

Knight, S. C., Xie, L., Deng, W., Guglielmi, B., Witkowsky, L. B., Bosanac, L., Zhang, E. T., Beheiry, M. E., Masson, J. B., Dahan, M., Liu, Z., Doudna, J. A., & Tjian, R. (2015). Dynamics of CRISPR-Cas9 genome interrogation in living cells. Science, 350(6262), 823–826. https://doi.org/10.1126/science.aac6572

Knott, G. J., & Doudna, J. A. (2018). CRISPR-Cas guides the future of genetic engineering. In Science (Vol. 361, Issue 6405, pp. 866–869). https://doi.org/10.1126/science.aat5011

Kok, F. O., Shin, M., Ni, C. W., Gupta, A., Grosse, A. S., VanImpel, A., Kirchmaier, B. C., Peterson-Maduro, J., Kourkoulis, G., Male, I., DeSantis, D. F., Sheppard-Tindell, S., Ebarasi, L., Betsholtz, C., Schulte-Merker, S., Wolfe, S. A., & Lawson, N. D. (2015). Reverse genetic screening reveals poor correlation between morpholino-induced and mutant phenotypes in zebrafish. Developmental Cell, 32(1), 97–108. https://doi.org/10.1016/j.devcel.2014.11.018

Kotani, H., Taimatsu, K., Ohga, R., Ota, S., & Kawahara, A. (2015a). Efficient multiple genome modifications induced by the crRNAs, tracrRNA and Cas9 protein complex in zebrafish. PLoS ONE, 10(5), 1–16. https://doi.org/10.1371/journal.pone.0128319

Kotani, H., Taimatsu, K., Ohga, R., Ota, S., & Kawahara, A. (2015b). Efficient multiple genome modifications induced by the crRNAs, tracrRNA and Cas9 protein complex in zebrafish. PLoS ONE, 10(5). https://doi.org/10.1371/journal.pone.0128319

Kozlowski, P. (2001). Combined SSCP/duplex analysis by capillary electrophoresis for more efficient mutation detection. Nucleic Acids Research, 29(14), 71e – 71. https://doi.org/10.1093/nar/29.14.e71

Kumar, M., Keller, B., Makalou, N., & Sutton, R. E. (2001). Systematic determination of the packaging limit of lentiviral vectors. Human Gene Therapy, 12(15), 1893–1905. https://doi.org/10.1089/104303401753153947

Lam, S. H., Chua, H. L., Gong, Z., Lam, T. J., & Sin, Y. M. (2004). Development and maturation of the immune system in zebrafish, Danio rerio: A gene expression profiling, in situ hybridization and immunological study. Developmental and Comparative Immunology, 28(1), 9–28. https://doi.org/10.1016/S0145-305X(03)00103-4

Lambert, C. J., Freshner, B. C., Chung, A., Stevenson, T. J., Bowles, D. M., Samuel, R., Gale, B. K., & Bonkowsky, J. L. (2018). An automated system for rapid cellular extraction from live zebrafish embryos and larvae: Development and application to genotyping. PLoS ONE, 13(3). https://doi.org/10.1371/journal.pone.0193180

Langheinrich, U., Vacun, G., & Wagner, T. (2003). Zebrafish embryos express an orthologue of HERG and are sensitive toward a range of QT-prolonging drugs inducing severe arrhythmia. Toxicology and Applied Pharmacology, 193(3), 370–382. https://doi.org/10.1016/j.taap.2003.07.012

Li, L., Wu, L. P., & Chandrasegaran, S. (1992). Functional domains in Fok I restriction endonuclease. Proceedings of the National Academy of Sciences of the United States of America, 89(10), 4275–4279. https://doi.org/10.1073/pnas.89.10.4275

Li, S. D., & Huang, L. (2006). Gene therapy progress and prospects: Non-viral gene therapy by systemic delivery. In Gene Therapy (Vol. 13, Issue 18, pp. 1313–1319). Nature Publishing Group. https://doi.org/10.1038/sj.gt.3302838

Li, Y., Li, Y., Cao, X., Jin, X., & Jin, T. (2017). Pattern recognition receptors in zebrafish provide functional and evolutionary insight into innate immune signaling pathways. In Cellular and Molecular Immunology (Vol. 14, Issue 1, pp. 80–89). Chinese Soc Immunology. https://doi.org/10.1038/cmi.2016.50

Lieber, M. R. (2010). The Mechanism of Double-Strand DNA Break Repair by the Nonhomologous DNA End-Joining Pathway. Annual Review of Biochemistry, 79(1), 181–211. https://doi.org/10.1146/annurev.biochem.052308.093131

Liu, C. X., Li, C. Y., Hu, C. C., Wang, Y., Lin, J., Jiang, Y. H., Li, Q., & Xu, X. (2018). CRISPR/Cas9-induced shank3b mutant zebrafish display autism-like behaviors. Molecular Autism, 9(1), 1–13. https://doi.org/10.1186/s13229-018-0204-x

Liu, K., Petree, C., Requena, T., Varshney, P., & Varshney, G. K. (2019). Expanding the CRISPR Toolbox in Zebrafish for Studying Development and Disease. Frontiers in Cell and Developmental Biology, 7(MAR). https://doi.org/10.3389/fcell.2019.00013

Liu, Q., Yuan, Y., Zhu, F., Hong, Y., & Ge, R. (2018). Efficient genome editing using CRISPR/Cas9 ribonucleoprotein approach in cultured Medaka fish cells. Biology Open, 7(8). https://doi.org/10.1242/bio.035170

Long, L., Guo, H., Yao, D., Xiong, K., Li, Y., Liu, P., Zhu, Z., & Liu, D. (2015). Regulation of transcriptionally active genes via the catalytically inactive Cas9 in C. elegans and D. rerio. In Cell Research (Vol. 25, Issue 5, pp. 638–641). Nature Publishing Group. https://doi.org/10.1038/cr.2015.35

Ma, Y., Zhang, L., & Huang, X. (2014). Genome modification by CRISPR/Cas9. FEBS Journal, 281(23), 5186–5193. https://doi.org/10.1111/febs.13110

Maeder, M. L., & Gersbach, C. A. (2016). Genome-editing technologies for gene and cell therapy. Molecular Therapy, 24(3), 430–446. https://doi.org/10.1038/mt.2016.10

Makarova, K. S., Wolf, Y. I., Iranzo, J., Shmakov, S. A., Alkhnbashi, O. S., Brouns, S. J. J., Charpentier, E., Cheng, D., Haft, D. H., Horvath, P., Moineau, S., Mojica, F. J. M., Scott, D., Shah, S. A., Siksnys, V., Terns, M. P., Venclovas, Č., White, M. F., Yakunin, A. F., … Koonin, E. V. (2020). Evolutionary classification of CRISPR–Cas systems: a burst of class 2 and derived variants. Nature Reviews Microbiology, 18(2), 67–83. https://doi.org/10.1038/s41579-019-0299-x

Mali, P., Aach, J., Stranges, B., Esvelt, K. M., Moosburner, M., Kosuri, S., Yang, L., & Church, G. M. (2013). CAs9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. https://doi.org/10.1038/nbt.2675

Marois, E., Van Den Ackerveken, G., & Bonas, U. (2002). The Xanthomonas type III effector protein AvrBs3 modulates plant gene expression and induces cell hypertrophy in the susceptible host. Molecular Plant-Microbe Interactions, 15(7), 637–646. https://doi.org/10.1094/MPMI.2002.15.7.637

Mashiko, D., Young, S. A. M., Muto, M., Kato, H., Nozawa, K., Ogawa, M., Noda, T., Kim, Y. J., Satouh, Y., Fujihara, Y., & Ikawa, M. (2014). Feasibility for a large scale mouse mutagenesis by injecting CRISPR/Cas plasmid into zygotes. Development Growth and Differentiation, 56(1), 122–129. https://doi.org/10.1111/dgd.12113

McCluskey, B. M., & Postlethwait, J. H. (2015). Phylogeny of zebrafish, a “model species,” within Danio, a “model genus.” Molecular Biology and Evolution, 32(3), 635–652. https://doi.org/10.1093/molbev/msu325

McMahon, J. M., Signori, E., Wells, K. E., Fazio, V. M., & Wells, D. J. (2001). Optimisation of electrotransfer of plasmid into skeletal muscle by pretreatment with hyaluronidase - Increased expression with reduced muscle damage. Gene Therapy, 8(16), 1264–1270. https://doi.org/10.1038/sj.gt.3301522

Meeker, N. D., Hutchinson, S. A., Ho, L., & Trede, N. S. (2007). Method for isolation of PCR-ready genomic DNA from zebrafish tissues. BioTechniques, 43(5), 610–614. https://doi.org/10.2144/000112619

Meijer, A. H., Gabby Krens, S. F., Medina Rodriguez, I. A., He, S., Bitter, W., Snaar-Jagalska, B. E., & Spaink, H. P. (2004). Expression analysis of the Toll-like receptor and TIR domain adaptor families of zebrafish. Molecular Immunology, 40(11), 773–783. https://doi.org/10.1016/j.molimm.2003.10.003

Mekler, V., Kuznedelov, K., & Severinov, K. (2020). Quantification of the affinities of CRISPR-Cas9 nucleases for cognate protospacer adjacent motif (PAM) sequences. J Biol Chem. https://doi.org/10.1074/jbc.RA119.012239

Men, W., Dong, Q., Shi, W., & Yao, K. (2019). Serotype distribution and antimicrobial resistance patterns of invasive pneumococcal disease isolates from children in mainland China—a systematic review. Brazilian Journal of Microbiology. https://doi.org/10.1007/s42770-019-00198-9

Meng, X., Noyes, M. B., Zhu, L. J., Lawson, N. D., & Wolfe, S. A. (2008). Targeted gene inactivation in zebrafish using engineered zinc-finger nucleases. Nature Biotechnology, 26(6), 695–701. https://doi.org/10.1038/nbt1398

Menke, A. L., Spitsbergen, J. M., Wolterbeek, A. P. M., & Woutersen, R. A. (2011). Normal Anatomy and Histology of the Adult Zebrafish. Toxicologic Pathology, 39(5), 759–775. https://doi.org/10.1177/0192623311409597

Meselson, M., & Yuan, R. (1968). DNA restriction enzyme from E. coli. Nature, 217(5134), 1110–1114. https://doi.org/10.1038/2171110a0

Miller, J. B., Zhang, S., Kos, P., Xiong, H., Zhou, K., Perelman, S. S., Zhu, H., & Siegwart, D. J. (2017). Non-Viral CRISPR/Cas Gene Editing In Vitro and In Vivo Enabled by Synthetic Nanoparticle Co-Delivery of Cas9 mRNA and sgRNA. Angewandte Chemie International Edition, 56(4), 1059–1063. https://doi.org/10.1002/anie.201610209

Miller, J. C., Tan, S., Qiao, G., Barlow, K. A., Wang, J., Xia, D. F., Meng, X., Paschon, D. E., Leung, E., Hinkley, S. J., Dulay, G. P., Hua, K. L., Ankoudinova, I., Cost, G. J., Urnov, F. D., Zhang, H. S., Holmes, M. C., Zhang, L., Gregory, P. D., & Rebar, E. J. (2011). A TALE nuclease architecture for efficient genome editing. Nature Biotechnology, 29(2), 143–150. https://doi.org/10.1038/nbt.1755

Miller, J., McLachlan, A. D., & Klug, A. (1985). Repetitive zinc-binding domains in the protein transcription factor IIIA from Xenopus oocytes. The EMBO Journal, 4(6), 1609–1614. https://doi.org/10.1002/j.1460-2075.1985.tb03825.x

Mirimin, L., O’Keeffe, D., Ruggiero, A., Bolton-Warberg, M., Vartia, S., & Fitzgerald, R. (2011). A quick, least-invasive, inexpensive and reliable method for sampling Gadus morhua postlarvae for genetic analysis. Journal of Fish Biology, 79(3), 801–805. https://doi.org/10.1111/j.1095-8649.2011.03049.x

Mojica, F. J. M., Díez-Villaseñor, C., Soria, E., & Juez, G. (2000). Biological significance of a family of regularly spaced repeats in the genomes of Archaea, Bacteria and mitochondria. In Molecular Microbiology (Vol. 36, Issue 1, pp. 244–246). https://doi.org/10.1046/j.1365-2958.2000.01838.x

Moscou, M. J., & Bogdanove, A. J. (2009). A simple cipher governs DNA recognition by TAL effectors. Science, 326(5959), 1501. https://doi.org/10.1126/science.1178817

Mout, R., Ray, M., Yesilbag Tonga, G., Lee, Y.-W., Tay, T., Sasaki, K., & Rotello, V. M. (2017). Direct Cytosolic Delivery of CRISPR/Cas9-Ribonucleoprotein for Efficient Gene Editing. ACS Nano, 11(3), 2452–2458. https://doi.org/10.1021/acsnano.6b07600

Mukherjee, A. S., & Beermann, W. (1965). Synthesis of Ribonucleic Acid by the X-Chromosomes of Drosophila melanogaster and the Problem of Dosage Compensation. Nature, 207(4998), 785–786. https://doi.org/10.1038/207785a0

Muller, H. J. (1932). Further studies on the nature and causes of gene mutations. In International Congress of Genetics (Vol. 6, Issue 1, pp. 213–255). http://darwin.amnh.org/bib/bibtexbrowser.php?key=muller1932yu83j&bib=Ev…;

Nagamine, C. M., Chan, K., & Lau, Y. F. (1989). A PCR artifact: generation of heteroduplexes. American Journal of Human Genetics, 47(1), 169–170. https://doi.org/10.1016/S0016-7878(61)80010-2

Nasevicius, A., & Ekker, S. C. (2000). Effective targeted gene “knockdown” in zebrafish. Nat Genet, 26(2), 216–220.

Neely, M. N., Pfeifer, J. D., & Caparon, M. (2002). Streptococcus-Zebrafish Model of Bacterial Pathogenesis. INFECTION AND IMMUNITY, 70(7), 3904–3914. https://doi.org/10.1128/IAI.70.7.3904-3914.2002

Nishimasu, H., Ran, F. A., Hsu, P. D., Konermann, S., Shehata, S. I., Dohmae, N., Ishitani, R., Zhang, F., & Nureki, O. (2014). Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell. https://doi.org/10.1016/j.cell.2014.02.001

Nobbs, A. H., Jenkinson, H. F., & Everett, D. B. (2015). Generic determinants of Streptococcus colonization and infection. Infection, Genetics and Evolution, 33, 361–370. https://doi.org/10.1016/j.meegid.2014.09.018

Novoa, B., & Figueras, A. (2012). Zebrafish: Model for the study of inflammation and the innate immune response to infectious diseases. Advances in Experimental Medicine and Biology, 946, 253–275. https://doi.org/10.1007/978-1-4614-0106-3_15

O’Callaghan, D., & Vergunst, A. (2010). Non-mammalian animal models to study infectious disease: worms or fly fishing? In Current Opinion in Microbiology (Vol. 13, Issue 1, pp. 79–85). Elsevier Current Trends. https://doi.org/10.1016/j.mib.2009.12.005

Ota, S., Hisano, Y., Ikawa, Y., & Kawahara, A. (2014). Multiple genome modifications by the CRISPR/Cas9 system in zebrafish. Genes to Cells, 19(7), 555–564. https://doi.org/10.1111/gtc.12154

Ota, S., Taimatsu, K., Yanagi, K., Namiki, T., Ohga, R., Higashijima, S. I., & Kawahara, A. (2016). Functional visualization and disruption of targeted genes using CRISPR/Cas9-mediated eGFP reporter integration in zebrafish. Scientific Reports, 6. https://doi.org/10.1038/srep34991

Ousterout, D. G., Perez-Pinera, P., Thakore, P. I., Kabadi, A. M., Brown, M. T., Qin, X., Fedrigo, O., Mouly, V., Tremblay, J. P., & Gersbach, C. A. (2013). Reading frame correction by targeted genome editing restores dystrophin expression in cells from duchenne muscular dystrophy patients. Molecular Therapy, 21(9), 1718–1726. https://doi.org/10.1038/mt.2013.111

Pallarès Masmitjà, M., Knödlseder, N., & Güell, M. (2019). CRISPR-gRNA Design. Methods in Molecular Biology, 1961, 3–11. https://doi.org/10.1007/978-1-4939-9170-9_1

Prykhozhij, S. V., Fuller, C., Steele, S. L., Veinotte, C. J., Razaghi, B., Robitaille, J. M., McMaster, C. R., Shlien, A., Malkin, D., & Berman, J. N. (2018). Optimized knock-in of point mutations in zebrafish using CRISPR/Cas9. Nucleic Acids Research, 46(17), 1–15. https://doi.org/10.1093/nar/gky512

Qi, L. S., Larson, M. H., Gilbert, L. A., Doudna, J. A., Weissman, J. S., Arkin, A. P., & Lim, W. A. (2013). Repurposing CRISPR as an RNA-Guided Platform for Sequence-Specific Control of Gene Expression. CELL, 152, 1173–1183. https://doi.org/10.1016/j.cell.2013.02.022

Ramaswamy, S., & Kordower, J. H. (2012). Gene therapy for Huntington’s disease. Neurobiology of Disease, 48(2), 243–254. https://doi.org/10.1016/j.nbd.2011.12.030

Ramirez, C. L., Foley, J. E., Wright, D. A., Müller-Lerch, F., Rahman, S. H., Cornu, T. I., Winfrey, R. J., Sander, J. D., Fu, F., Townsend, J. A., Cathomen, T., Voytas, D. F., & Joung, J. K. (2008). Unexpected failure rates for modular assembly of engineered zinc fingers (Nature Methods (2008) vol. 5 (374-375)). Nature Methods, 5(6), 575. https://doi.org/10.1038/nmeth0608-575a

Ran, F. A., Cong, L., Yan, W. X., Scott, D. A., Gootenberg, J. S., Kriz, A. J., Zetsche, B., Shalem, O., Wu, X., Makarova, K. S., Koonin, E. V., Sharp, P. A., & Zhang, F. (2015). In vivo genome editing using Staphylococcus aureus Cas9. Nature, 520(7546), 186–191. https://doi.org/10.1038/nature14299

Rath, D., Amlinger, L., Rath, A., & Lundgren, M. (2015). The CRISPR-Cas immune system: Biology, mechanisms and applications. In Biochimie. https://doi.org/10.1016/j.biochi.2015.03.025

Renshaw, S. A., Loynes, C. A., Trushell, D. M. I., Elworthy, S., Ingham, P. W., & Whyte, M. K. B. (2006). A transgenic zebrafish model of neutrophilic inflammation. Blood, 108(13), 3976–3978. https://doi.org/10.1182/blood-2006-05-024075

Richardson, C. D., Ray, G. J., DeWitt, M. A., Curie, G. L., & Corn, J. E. (2016). Enhancing homology-directed genome editing by catalytically active and inactive CRISPR-Cas9 using asymmetric donor DNA. Nature Biotechnology, 34(3), 339–344. https://doi.org/10.1038/nbt.3481

Rounioja, S., Saralahti, A., Rantala, L., Parikka, M., Henriques-Normark, B., Silvennoinen, O., & Rämet, M. (2012). Defense of zebrafish embryos against Streptococcus pneumoniae infection is dependent on the phagocytic activity of leukocytes. Developmental and Comparative Immunology, 36(2), 342–348. https://doi.org/10.1016/j.dci.2011.05.008

Rudin, N., Sugarman, E., & Haber, J. E. (1989). Genetic and physical analysis of double-strand break repair and recombination in Saccharomyces cerevisiae. Genetics, 122(3), 519–534.

Ryan, R. P., Vorhölter, F.-J., Potnis, N., Jones, J. B., Van Sluys, M.-A., Bogdanove, A. J., & Dow, J. M. (2011). Pathogenomics of Xanthomonas: understanding bacterium–plant interactions. Nature Reviews Microbiology, 9(5), 344–355. https://doi.org/10.1038/nrmicro2558

Sander, J. D., Cade, L., Khayter, C., Reyon, D., Peterson, R. T., Joung, J. K., & Yeh, J. J. (2011). Targeted gene disruption in somatic zebrafish cells using engineered TALENs. Nature Biotechnology, 29(8), 697–698. https://doi.org/10.1038/nbt.1934

Sapranauskas, R., Gasiunas, G., Fremaux, C., Barrangou, R., Horvath, P., & Siksnys, V. (n.d.). The Streptococcus thermophilus CRISPR/Cas system provides immunity in Escherichia coli. https://doi.org/10.1093/nar/gkr606

Saralahti, A., Piippo, H., Parikka, M., Henriques-Normark, B., Rämet, M., & Rounioja, S. (2014). Adult zebrafish model for pneumococcal pathogenesis. Developmental and Comparative Immunology, 42(2), 345–353. https://doi.org/10.1016/j.dci.2013.09.009

Scherer, S., & Davis, R. W. (1979). Replacement of chromosome segments with altered DNA sequences constructed in vitro. Proceedings of the National Academy of Sciences of the United States of America, 76(10), 4951–4955. https://doi.org/10.1073/pnas.76.10.4951

Schmid, B., Hruscha, A., Hogl, S., Banzhaf-Strathmann, J., Strecker, K., Van Der Zee, J., Teucke, M., Eimer, S., Hegermann, J., Kittelmann, M., Kremmer, E., Cruts, M., Solchenberger, B., Hasenkamp, L., Van Bebber, F., Van Broeckhoven, C., Edbauer, D., Lichtenthaler, S. F., & Haass, C. (2013). Loss of ALS-associated TDP-43 in zebrafish causes muscle degeneration, vascular dysfunction, and reduced motor neuron axon outgrowth. Proceedings of the National Academy of Sciences of the United States of America, 110(13), 4986–4991. https://doi.org/10.1073/pnas.1218311110

Schmidt, F., & Grimm, D. (2015). CRISPR genome engineering and viral gene delivery: A case of mutual attraction. Biotechnology Journal, 10(2), 258–272. https://doi.org/10.1002/biot.201400529

Schulte-Merker, S., & Stainier, D. Y. R. (2014). Out with the old, in with the new: Reassessing morpholino knockdowns in light of genome editing technology. Development (Cambridge), 141(16), 3103–3104. https://doi.org/10.1242/dev.112003

Segal, D. J., Beerli, R. R., Blancafort, P., Dreier, B., Effertz, K., Huber, A., Koksch, B., Lund, C. V., Magnenat, L., Valente, D., & Barbas, C. F. (2003). Evaluation of a modular strategy for the construction of novel polydactyl zinc finger DNA-binding proteins. Biochemistry, 42(7), 2137–2148. https://doi.org/10.1021/bi026806o

Senís, E., Fatouros, C., Große, S., Wiedtke, E., Niopek, D., Mueller, A. K., Börner, K., & Grimm, D. (2014). CRISPR/Cas9-mediated genome engineering: An adeno-associated viral (AAV) vector toolbox. Biotechnology Journal, 9(11), 1402–1412. https://doi.org/10.1002/biot.201400046

Shah, A. N., Davey, C. F., Whitebirch, A. C., Miller, A. C., & Moens, C. B. (2016). Rapid Reverse Genetic Screening Using CRISPR in Zebrafish. Zebrafish, 13(2), 152–153. https://doi.org/10.1089/zeb.2015.29000.sha

Simpson, R. J., Wilding, C. S., & Grahame, J. (1999). Simple protocol for extracting nuclear DNA from single embryos of a marine snail. BioTechniques, 26(6), 1050–1052. https://doi.org/10.2144/99266bm08

Smith, H. O., & Welcox, K. W. (1970). A Restriction enzyme from Hemophilus influenzae. I. Purification and general properties. Journal of Molecular Biology, 51(2), 379–391. https://doi.org/10.1016/0022-2836(70)90149-X

Smithies, O., Gregg, R. G., Boggs, S. S., Koralewski, M. A., & Kucherlapati, R. S. (1985). Insertion of DNA sequences into the human chromosomal β-globin locus by homologous recombination. Nature, 317(6034), 230–234. https://doi.org/10.1038/317230a0

Sprague, J. (2006). The Zebrafish Information Network: the zebrafish model organism database. Nucleic Acids Research, 34(90001), D581–D585. https://doi.org/10.1093/nar/gkj086

Stainier, D. Y. R., Raz, E., Lawson, N. D., Ekker, S. C., Burdine, R. D., Eisen, J. S., Ingham, P. W., Schulte-Merker, S., Yelon, D., Weinstein, B. M., Mullins, M. C., Wilson, S. W., Ramakrishnan, L., Amacher, S. L., Neuhauss, S. C. F., Meng, A., Mochizuki, N., Panula, P., & Moens, C. B. (2017). Guidelines for morpholino use in zebrafish. PLOS Genetics, 13(10), e1007000. https://doi.org/10.1371/journal.pgen.1007000

Sternberg, S. H., Richter, H., Charpentier, E., & Qimron, U. (2016). Adaptation in CRISPR-Cas Systems. Molecular Cell, 61(6), 797–808. https://doi.org/10.1016/j.molcel.2016.01.030

Sullivan, C., & Kim, C. H. (2008). Zebrafish as a model for infectious disease and immune function. Fish and Shellfish Immunology, 25(4), 341–350. https://doi.org/10.1016/j.fsi.2008.05.005

Summerton, J. (2007). Morpholino, siRNA, and S-DNA Compared: Impact of Structure and Mechanism of Action on Off-Target Effects and Sequence Specificity. Current Topics in Medicinal Chemistry, 7(7), 651–660. https://doi.org/10.2174/156802607780487740

Summerton, J., & Weller, D. (1997). Morpholino Antisense Oligomers: Design, Preparation, and Properties. Antisense and Nucleic Acid Drug Development, 7(3), 187–195. https://doi.org/10.1089/oli.1.1997.7.187

Sun, Z. (2004). A genetic screen in zebrafish identifies cilia genes as a principal cause of cystic kidney. Development, 131(16), 4085–4093. https://doi.org/10.1242/dev.01240

Sung, Y. H., Kim, J. M., Kim, H. T., Lee, J., Jeon, J., Jin, Y., Choi, J. H., Ban, Y. H., Ha, S. J., Kim, C. H., Lee, H. W., & Kim, J. S. (2014). Highly efficient gene knockout in mice and zebrafish with RNA-guided endonucleases. Genome Research, 24(1), 125–131. https://doi.org/10.1101/gr.163394.113

Szczepek, M., Brondani, V., Büchel, J., Serrano, L., Segal, D. J., & Cathomen, T. (2007). Structure-based redesign of the dimerization interface reduces the toxicity of zinc-finger nucleases. Nature Biotechnology, 25(7), 786–793. https://doi.org/10.1038/nbt1317

Tampakaki, A. P., Fadouloglou, V. E., Gazi, A. D., Panopoulos, N. J., & Kokkinidis, M. (2004). Conserved features of type III secretion. Cellular Microbiology, 6(9), 805–816. https://doi.org/10.1111/j.1462-5822.2004.00432.x

Tautz, D. (1992). Problems and paradigms: Redundancies, development and the flow of information. BioEssays, 14(4), 263–266. https://doi.org/10.1002/bies.950140410

Terns, M. P., & Terns, R. M. (2012). CRISPR-Based Adaptive Immune Systems. Bone, 23(1), 1–7. https://doi.org/10.1038/jid.2014.371

Thakore, P. I., D’Ippolito, A. M., Song, L., Safi, A., Shivakumar, N. K., Kabadi, A. M., Reddy, T. E., Crawford, G. E., & Gersbach, C. A. (2015). Highly specific epigenome editing by CRISPR-Cas9 repressors for silencing of distal regulatory elements. Nature Methods, 12(12), 1143–1149. https://doi.org/10.1038/nmeth.3630

Tobin, D. M., May, R. C., & Wheeler, R. T. (2012). Zebrafish: A see-through host and a fluorescent toolbox to probe host-pathogen interaction. PLoS Pathogens, 8(1), 8–10. https://doi.org/10.1371/journal.ppat.1002349

Tobin, D. M., Vary, J. C., Ray, J. P., Walsh, G. S., Dunstan, S. J., Bang, N. D., Hagge, D. A., Khadge, S., King, M. C., Hawn, T. R., Moens, C. B., & Ramakrishnan, L. (2010). The lta4h Locus Modulates Susceptibility to Mycobacterial Infection in Zebrafish and Humans. Cell, 140(5), 717–730. https://doi.org/10.1016/j.cell.2010.02.013

Torraca, V., & Mostowy, S. (2018). Zebrafish Infection: From Pathogenesis to Cell Biology. Trends in Cell Biology, 28(2), 143–156. https://doi.org/10.1016/j.tcb.2017.10.002

Trede, N. S., Langenau, D. M., Traver, D., Look, A. T., & Zon, L. I. (2004). The use of zebrafish to understand immunity. Immunity, 20(4), 367–379. https://doi.org/10.1016/S1074-7613(04)00084-6

Tsai, S. Q., Wyvekens, N., Khayter, C., Foden, J. A., Thapar, V., Reyon, D., Goodwin, M. J., Aryee, M. J., & Joung, J. K. (2014). Dimeric CRISPR RNA-guided FokI nucleases for highly specific genome editing. Nature Biotechnology, 32(6), 569–576. https://doi.org/10.1038/nbt.2908

Turner, C. R., Uy, K. L., & Everhart, R. C. (2015). Fish environmental DNA is more concentrated in aquatic sediments than surface water. Biological Conservation, 183, 93–102. https://doi.org/10.1016/j.biocon.2014.11.017

Urnov, F. D., Rebar, E. J., Holmes, M. C., Zhang, H. S., & Gregory, P. D. (2010). Genome editing with engineered zinc finger nucleases. Nature Reviews Genetics, 11(9), 636–646. https://doi.org/10.1038/nrg2842

van der Oost, J., Jore, M. M., Westra, E. R., Lundgren, M., & Brouns, S. J. J. (2009). CRISPR-based adaptive and heritable immunity in prokaryotes. Trends in Biochemical Sciences, 34(8), 401–407. https://doi.org/10.1016/j.tibs.2009.05.002

Vargas, R. A., Sarmiento, K., & Vásquez, I. C. (2015). Zebrafish (Danio rerio): A Potential Model for Toxinological Studies. Zebrafish, 12(5), 320–326. https://doi.org/10.1089/zeb.2015.1102

Villamizar, N., Ribas, L., Piferrer, F., Vera, L. M., & Sánchez-Vázquez, F. J. (2012). Impact of Daily Thermocycles on Hatching Rhythms, Larval Performance and Sex Differentiation of Zebrafish. PLoS ONE, 7(12), e52153. https://doi.org/10.1371/journal.pone.0052153

Villarreal, M. A., Biediger, N. M., Bonner, N. A., Miller, J. N., Zepeda, S. K., Ricard, B. J., García, D. M., & Lewis, K. A. (2017). Determining Zebrafish Epitope Reactivity to Commercially Available Antibodies. Zebrafish, 14(4), 387–389. https://doi.org/10.1089/zeb.2016.1401

Vishwanath, W. (2010). Danio rerio. https://www.iucnredlist.org/species/166487/6219667

Watson, J. D., & Crick, F. H. (1953). The structure of DNA. Cold Spring Harbor Symposia on Quantitative Biology, 18, 123–131. https://doi.org/10.1101/SQB.1953.018.01.020

Wells, D. J. (2004). Gene therapy progress and prospects: Electroporation and other physical methods. In Gene Therapy (Vol. 11, Issue 18, pp. 1363–1369). Nature Publishing Group. https://doi.org/10.1038/sj.gt.3302337

WHO. (2013). WHO | Estimated Hib and pneumococcal deaths for children under 5 years of age, 2008. World Health Organization. https://www.who.int/immunization/monitoring_surveillance/burden/estimat…

Wigler, M., Silverstein, S., Lee, L. S., Pellicer, A., Cheng, Y. chi, & Axel, R. (1977). Transfer of purified herpes virus thymidine kinase gene to cultured mouse cells. Cell, 11(1), 223–232. https://doi.org/10.1016/0092-8674(77)90333-6

Wilkinson, R. N., Elworthy, S., Ingham, P. W., & van Eeden, F. J. M. (2013). A method for high-throughput PCR-based genotyping of larval zebrafish tail biopsies. BioTechniques, 55(6), 314–316. https://doi.org/10.2144/000114116

Wolfe, S. A., Nekludova, L., & Pabo, C. O. (2009). DNA recognition by Cys2His2 zinc finger proteins. Biophysical Structure, 25(1), 22–29. https://doi.org/10.1093/bioinformatics/btn580

Woods, I. G., Kelly, P. D., Chu, F., Ngo-Hazelett, P., Yan, Y.-L., Huang, H., Postlethwait, J. H., & Talbot, W. S. (2000). A Comparative Map of the Zebrafish Genome. Genome Research, 10(12), 1903–1914. https://doi.org/10.1101/gr.10.12.1903

Wu, Z., Yang, H., & Colosi, P. (2010). Effect of Genome Size on AAV Vector Packaging. Molecular Therapy, 18(1), 80–86. https://doi.org/10.1038/mt.2009.255

Xiao, A., Wang, Z., Hu, Y., Wu, Y., Luo, Z., Yang, Z., Zu, Y., Li, W., Huang, P., Tong, X., Zhu, Z., Lin, S., & Zhang, B. (2013). Chromosomal deletions and inversions mediated by TALENs and CRISPR/Cas in zebrafish. Nucleic Acids Research, 41(14), 1–11. https://doi.org/10.1093/nar/gkt464

Xing, L., Quist, T. S., Stevenson, T. J., Dahlem, T. J., & Bonkowsky, J. L. (2014). Rapid and efficient zebrafish genotyping using PCR with high-resolution melt analysis. Journal of Visualized Experiments, 84. https://doi.org/10.3791/51138

Yue, G. H., & Orban, L. (2001). Rapid isolation of DNA from fresh and preserved fish scales for polymerase chain reaction. Marine Biotechnology, 3(3), 199–204. https://doi.org/10.1007/s10126-001-0010-9

ZFIN The Zebrafish Information Network. (n.d.). Retrieved March 22, 2020, from https://zfin.org/

Zhang, F., Abudayyeh, O. O., Gootenberg, J. S., Sciences, C., & Mathers, L. (2020). A protocol for detection of COVID-19 using CRISPR diagnostics. Bioarchive, 1–8.

Zilberman, N., Reikhav, S., Hulata, G., & Ron, M. (2006). High-throughput genomic DNA extraction protocol from tilapia’s fin tissue. Aquaculture, 255(1–4), 597–599. https://doi.org/10.1016/j.aquaculture.2005.11.042

Zon, L. I., & Peterson, R. T. (2005). In vivo drug discovery in the zebrafish. Nature Reviews Drug Discovery, 4(1), 35–44. https://doi.org/10.1038/nrd1606

Zou, M., Friedrich, R. W., & Bianco, I. H. (2016). Targeted Electroporation in Embryonic, Larval, and Adult Zebrafish (K. Kawakami, E. E. Patton, & M. Orger (Eds.); Vol. 1451, pp. 259–269). Springer New York. https://doi.org/10.1007/978-1-4939-3771-4_17

 

Download scriptie (1001.49 KB)
Universiteit of Hogeschool
Odisee
Thesis jaar
2020
Promotor(en)
Dr. Anni Saralahti