New Wine in Old Wineskin: Progressive Development of Genomic Editing in Bombyx mori

Moses A. Nartey, Li Muwang, Li Tao, Daniel Adjibolosoo

Abstract


The Bombyx mori insect emerging from lepidopteran family is exploited as an important remunerative specie, vis-a-vis as a model organism for scientific findings. It is unequivocally a primal insect model in which genomic altering tool was used to establish a gene function in vitro. A significant milestone in Bombyx genetics is evidenced by contemporary advancement in germline transformation and whole-genome sequencing, producing genetically engineered silkworms (new wine in an old wineskin) to generate recombinant silk proteins for biomedical and industrial applications. Here, we mainly discuss the contemporary advancements in genome modification approaches employed in silkworm research focusing on the three genomic engineering methodologies (ZFNs, TALENs, and CRISPR/Cas9). Since the inception of genomic editing using ZFNs, few experiments under Bombyx have been recorded. The mutation rates in Bombyx using ZFNs are relatively low compared to those using TALENs techniques. TALENs exhibit much simpler design as a single TALE and recognize one nucleotide, unlike ZFNs, which recognize three nucleotides. CRISPR/Cas9 offers flexibility, as well as easy multiplexing and scaling out of reach of the performances of ZFNs and TALENs.

 

Keywords: Targeted mutagenesis, homologous recombination, clustered regularly interspaced palindromic repeats, transcription activator-like effectors nucleases, and zinc finger nucleases

Cite this Article

Nartey Moses A, Li Muwang, Li Tao et al. New Wine in Old Wineskin: Progressive Development of Genomic Editing in Bombyx mori. Research & Reviews: Journal of Agricultural Science and Technology. 2018; 7(3): 17–25p.


Keywords


targeted mutagenesis, homologous recombination, clustered regularly interspaced palindromic repeats, transcription activator-like effectors nucleases, and zinc finger nucleases

References


Kawarabata, T., Biological studies on the ancestral insect species of the silkworm, Bombyx mori. Report for a Scientific Research Supported by a Grant-in-Aid from the Ministry of Education, Science and Culture of Japan; a Separate Issue, 1998.

Tazima, Y., The silkworm: an important laboratory tool. 1978.

Goldsmith, M.R., T. Shimada, and H. Abe, The genetics and genomics of the silkworm, Bombyx mori. Annu. Rev. Entomol., 2005. 50: p. 71-100.

Consortium, I.S.G., The genome of a lepidopteran model insect, the silkworm Bombyx mori. Insect biochemistry and molecular biology, 2008. 38(12): p. 1036-1045.

Xia, Q., et al., A draft sequence for the genome of the domesticated silkworm (Bombyx mori). Science, 2004. 306(5703): p. 1937-1940.

Tamura, T., et al., Germline transformation of the silkworm Bombyx mori L. using a piggyBac transposon-derived vector. Nature biotechnology, 2000. 18(1): p. 81.

Imamura, M., et al., Targeted gene expression using the GAL4/UAS system in the silkworm Bombyx mori. Genetics, 2003. 165(3): p. 1329-1340.

Uchino, K., et al., Construction of a piggyBac-based enhancer trap system for the analysis of gene function in silkworm Bombyx mori. Insect biochemistry and molecular biology, 2008. 38(12): p. 1165-1173.

Long, D.-P., et al., FLP recombinase-mediated site-specific recombination in silkworm, Bombyx mori. PLoS One, 2012. 7(6): p. e40150.

Galetto, R., P. Duchateau, and F. Pâques, Targeted approaches for gene therapy and the emergence of engineered meganucleases. Expert opinion on biological therapy, 2009. 9(10): p. 1289-1303.

Smith, J., et al., A combinatorial approach to create artificial homing endonucleases cleaving chosen sequences. Nucleic acids research, 2006. 34(22): p. e149-e149.

Urnov, F.D., et al., Genome editing with engineered zinc finger nucleases. Nature Reviews Genetics, 2010. 11(9): p. 636.

Miller, J., et al., Leung E, Hinkley SJ, Dulay GP, Hua KL, Ankoudinova I, Cost GJ, Urnov FD, 11 Zhang HS, Holmes MC, Zhang L, Gregory PD, Rebar EJ. A TALE nuclease. 12: p. 143-148.

Boch, J., et al., Breaking the code of DNA binding specificity of TAL-type III effectors. Science, 2009. 326(5959): p. 1509-1512.

Moscou, M.J. and A.J. Bogdanove, A simple cipher governs DNA recognition by TAL effectors. Science, 2009. 326(5959): p. 1501-1501.

Joung, J.K. and J.D. Sander, TALENs: a widely applicable technology for targeted genome editing. Nature reviews Molecular cell biology, 2013. 14(1): p. 49.

Horvath, P. and R. Barrangou, CRISPR/Cas, the immune system of bacteria and archaea. Science, 2010. 327(5962): p. 167-170.

Jinek, M., et al., A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. science, 2012: p. 1225829.

Cho, S.W., et al., Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nature biotechnology, 2013. 31(3): p. 230.

Hwang, W.Y., et al., Heritable and precise zebrafish genome editing using a CRISPR-Cas system. PloS one, 2013. 8(7): p. e68708.

Hwang, W.Y., et al., Efficient genome editing in zebrafish using a CRISPR-Cas system. Nature biotechnology, 2013. 31(3): p. 227.

Jiang, W., et al., RNA-guided editing of bacterial genomes using CRISPR-Cas systems. Nature biotechnology, 2013. 31(3): p. 233.

Takasu, Y., et al., Targeted mutagenesis in the silkworm Bombyx mori using zinc finger nuclease mRNA injection. Insect biochemistry and molecular biology, 2010. 40(10): p. 759-765.

Sajwan, S., et al., Efficient disruption of endogenous Bombyx gene by TAL effector nucleases. Insect biochemistry and molecular biology, 2013. 43(1): p. 17-23.

Ma, S., et al., Highly efficient and specific genome editing in silkworm using custom TALENs. PloS one, 2012. 7(9): p. e45035.

Takasu, Y., et al., Efficient TALEN construction for Bombyx mori gene targeting. PLoS One, 2013. 8(9): p. e73458.

Klug, A., The discovery of zinc fingers and their applications in gene regulation and genome manipulation. Annual review of biochemistry, 2010. 79: p. 213-231.

Smith, J., et al., Requirements for double-strand cleavage by chimeric restriction enzymes with zinc finger DNA-recognition domains. Nucleic acids research, 2000. 28(17): p. 3361-3369.

Ly, J.P., T. Onay, and S.E. Quaggin, Mouse models to study kidney development, function and disease. Current opinion in nephrology and hypertension, 2011. 20(4): p. 382-390.

Urnov, F.D., et al., Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature, 2005. 435(7042): p. 646.

Ramirez, C.L., et al., Unexpected failure rates for modular assembly of engineered zinc fingers. Nature methods, 2008. 5(5): p. 374.

Kim, J.-S., H.J. Lee, and D. Carroll, Genome editing with modularly assembled zinc-finger nucleases. Nat. Methods, 2010. 7(91).

Doyon, Y., et al., Enhancing zinc-finger-nuclease activity with improved obligate heterodimeric architectures. Nature methods, 2011. 8(1): p. 74.

Meng, X., et al., Profiling the DNA-binding specificities of engineered Cys2His2 zinc finger domains using a rapid cell-based method. Nucleic acids research, 2007. 35(11): p. e81.

Foley, J.E., et al., Rapid mutation of endogenous zebrafish genes using zinc finger nucleases made by Oligomerized Pool ENgineering (OPEN). PloS one, 2009. 4(2): p. e4348.

Quan, G., T. Kanda, and T. Tamura, Induction of the white egg 3 mutant phenotype by injection of the double‐stranded RNA of the silkworm white gene. Insect molecular biology, 2002. 11(3): p. 217-222.

Kômoto, N., et al., A single-base deletion in an ABC transporter gene causes white eyes, white eggs, and translucent larval skin in the silkworm w-3oe mutant. Insect biochemistry and molecular biology, 2009. 39(2): p. 152-156.

Fujii, T., et al., Transgenic analysis of the BmBLOS2 gene that governs the translucency of the larval integument of the silkworm, Bombyx mori. Insect molecular biology, 2010. 19(5): p. 659-667.

Carroll, D., Genome engineering with zinc-finger nucleases. Genetics, 2011. 188(4): p. 773-782.

Boch, J., TALEs of genome targeting. Nature biotechnology, 2011. 29(2): p. 135.

Christian, M., et al., Targeting DNA double-strand breaks with TAL effector nucleases. Genetics, 2010. 186(2): p. 757-761.

Daimon, T., T. Kiuchi, and Y. Takasu, Recent progress in genome engineering techniques in the silkworm, Bombyx mori. Development, growth & differentiation, 2014. 56(1): p. 14-25.

Xiao, A., et al., EENdb: a database and knowledge base of ZFNs and TALENs for endonuclease engineering. Nucleic acids research, 2012. 41(D1): p. D415-D422.

Liu, J., et al., Efficient and specific modifications of the Drosophila genome by means of an easy TALEN strategy. Journal of genetics and genomics, 2012. 39(5): p. 209-215.

Katsuyama, T., et al., An efficient strategy for TALEN-mediated genome engineering in Drosophila. Nucleic acids research, 2013. 41(17): p. e163-e163.

Beumer, K.J., et al., Comparing zinc finger nucleases and transcription activator-like effector nucleases for gene targeting in Drosophila. G3: Genes, Genomes, Genetics, 2013. 3(10): p. 1717-1725.

Cermak, T., et al., Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic acids research, 2011. 39(12): p. e82-e82.

Wang, Y., et al., Site-specific, TALENs-mediated transformation of Bombyx mori. Insect biochemistry and molecular biology, 2014. 55: p. 26-30.

Xu, J., et al., Transcription activator‐like effector nuclease (TALEN)‐mediated female‐specific sterility in the silkworm, Bombyx mori. Insect molecular biology, 2014. 23(6): p. 800-807.

Suzuki, M.G., et al., Analysis of the biological functions of a doublesex homologue in Bombyx mori. Development genes and evolution, 2003. 213(7): p. 345-354.

Ishino, Y., et al., Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product. Journal of bacteriology, 1987. 169(12): p. 5429-5433.

Barrangou, R., et al., CRISPR provides acquired resistance against viruses in prokaryotes. Science, 2007. 315(5819): p. 1709-1712.

Sorek, R., C.M. Lawrence, and B. Wiedenheft, CRISPR-mediated adaptive immune systems in bacteria and archaea. Annual review of biochemistry, 2013. 82: p. 237-266.

Mali, P., et al., RNA-guided human genome engineering via Cas9. Science, 2013. 339(6121): p. 823-826.

Makarova, K.S., et al., An updated evolutionary classification of CRISPR–Cas systems. Nature Reviews Microbiology, 2015. 13(11): p. 722.

Wang, Y., et al., The CRISPR/Cas system mediates efficient genome engineering in Bombyx mori. Cell research, 2013. 23(12): p. 1414.

Liu, Y., et al., Highly efficient multiplex targeted mutagenesis and genomic structure variation in Bombyx mori cells using CRISPR/Cas9. Insect biochemistry and molecular biology, 2014. 49: p. 35-42.

Wei, W., et al., Heritable genome editing with CRISPR/Cas9 in the silkworm, Bombyx mori. PloS one, 2014. 9(7): p. e101210.

Ma, S., et al., CRISPR/Cas9 mediated multiplex genome editing and heritable mutagenesis of BmKu70 in Bombyx mori. Scientific reports, 2014. 4: p. 4489.

Zhang, Z., et al., Functional analysis of Bombyx Wnt1 during embryogenesis using the CRISPR/Cas9 system. Journal of insect physiology, 2015. 79: p. 73-79.

Zhang, L. and R.D. Reed, Genome editing in butterflies reveals that spalt promotes and Distal-less represses eyespot colour patterns. Nature communications, 2016. 7: p. 11769.

Perry, M., et al., Molecular logic behind the three-way stochastic choices that expand butterfly colour vision. Nature, 2016. 535(7611): p. 280.

Xu, J., et al., Bombyx mori P-element Somatic Inhibitor (BmPSI) is a key auxiliary factor for silkworm male sex determination. PLoS genetics, 2017. 13(1): p. e1006576.

Liu, Q., et al., Deletion of the Bombyx mori odorant receptor co-receptor (BmOrco) impairs olfactory sensitivity in silkworms. Insect biochemistry and molecular biology, 2017. 86: p. 58-67.

Xin, H.h., et al., Transcription factor Bmsage plays a crucial role in silk gland generation in silkworm, Bombyx mori. Archives of insect biochemistry and physiology, 2015. 90(2): p. 59-69.

Liao, H.-K., et al., Use of the CRISPR/Cas9 system as an intracellular defense against HIV-1 infection in human cells. Nature communications, 2015. 6: p. 6413.

Hongbao, M., et al., CRISPR and Cancer Biology Research Literatures.

Dong, Z.-Q., et al., Establishment of a highly efficient virus-inducible CRISPR/Cas9 system in insect cells. Antiviral research, 2016. 130: p. 50-57.

Dong, Z., et al., Excision of Nucleopolyhedrovirus Form Transgenic Silkworm Using the CRISPR/Cas9 System. Frontiers in Microbiology, 2018. 9: p. 209.

Dong, Z.-Q., et al., Baculovirus LEF-11 Hijack Host ATPase ATAD3A to Promote Virus Multiplication in Bombyx mori cells. Scientific Reports, 2017. 7: p. 46187.

Chen, S., et al., Transgenic CRISPR/Cas9-mediated viral gene targeting for antiviral therapy of Bombyx mori nucleopolyhedrovirus (BmNPV). Journal of Virology, 2017: p. JVI. 02465-16.

Jindra, M., S.R. Palli, and L.M. Riddiford, The juvenile hormone signaling pathway in insect development. Annual review of entomology, 2013. 58: p. 181-204.

Riddiford, L.M., Juvenile hormone: the status of its “status quo” action. Archives of insect biochemistry and physiology, 1996. 32(3‐4): p. 271-286.

Zhang, Z., et al., Depletion of juvenile hormone esterase extends larval growth in Bombyx mori. Insect biochemistry and molecular biology, 2017. 81: p. 72-79.

Zeng, B., et al., The FOXO transcription factor controls insect growth and development by regulating juvenile hormone degradation in the silkworm, Bombyx mori. Journal of Biological Chemistry, 2017. 292(28): p. 11659-11669.

Ling, L., et al., MiR-2 family targets awd and fng to regulate wing morphogenesis in Bombyx mori. RNA biology, 2015. 12(7): p. 742-748.

Daimon, T., et al., Knockout silkworms reveal a dispensable role for juvenile hormones in holometabolous life cycle. Proceedings of the National Academy of Sciences, 2015. 112(31): p. E4226-E4235.

Li, Z., et al., Ectopic expression of ecdysone oxidase impairs tissue degeneration in Bombyx mori. Proc. R. Soc. B, 2015. 282(1809): p. 20150513.

Zhu, L., et al., CRISPR/Cas9-mediated knockout of factors in non-homologous end joining pathway enhances gene targeting in silkworm cells. Scientific reports, 2015. 5: p. 18103.

Zeng, B., et al., Expansion of CRISPR targeting sites in Bombyx mori. Insect biochemistry and molecular biology, 2016. 72: p. 31-40.




DOI: https://doi.org/10.37591/rrjoast.v7i3.1239

Refbacks



Copyright (c) 2018 Research & Reviews: Journal of Agricultural Science and Technology