2,862
Views
12
CrossRef citations to date
0
Altmetric
Review

Random mutagenesis and precise gene editing technologies: applications in algal crop improvement and functional genomics

ORCID Icon &
Pages 466-481 | Received 12 May 2017, Accepted 17 Jul 2017, Published online: 10 Oct 2017

References

  • Abdallah, N.A., Prakash, C.S. & McHughen, A.G. (2015). Genome editing for crop improvement: challenges and opportunities. GM Crops & Food, 6: 183–205.
  • Abomohra, A.E-F., El-Sheekh, M. & Hanelt, D. (2016). Protoplast fusion and genetic recombination between Ochromonas danica (Chrysophyta) and Haematococcus pluvialis (Chlorophyta). Phycologia, 55: 65–71.
  • Adam, M., Lentz, K.E. & Loppes, R. (1993). Insertional mutagenesis to isolate acetate-requiring mutants in Chlamydomonas reinhardtii. FEMS Microbiology Letters, 110: 265–268.
  • Al-Ahmad, H., Dwyer, J., Moloney, M. & Gressel, J. (2006). Mitigation of establishment of Brassica napus transgenes in volunteers using a tandem construct containing a selectively unfit gene. Plant Biotechnology Journal, 4: 7–21.
  • Anila, N., Chandrashekar, A., Ravishankar, G.A. & Sarada, R. (2011). Establishment of Agrobacterium tumefaciens-mediated genetic transformation in Dunaliella bardawil. European Journal of Phycology, 46: 36–44.
  • Apt, K.E., Kroth-Pancic, P.G. & Grossman, A.R. (1996). Stable nuclear transformation of the diatom Phaeodactylum tricornutum. Molecular and General Genetics, 252: 572–579.
  • Bae, S., Park, J. & Kim, J.S. (2014). Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinformatics, 30: 1473–1475.
  • Batchvarov, S. (1993). Garlic Allium sativum L. In Genetic Improvement of Vegetable Crops (Kalloo, G. & Bergh, B.O., editors), 15–27. Pergamon Press, New York.
  • Bateman, J.M. & Purton, S. (2000). Tools for chloroplast transformation in Chlamydomonas: expression vectors and a new dominant selectable marker. Molecular and General Genetics, 263: 404–410.
  • 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: 10570–10575.
  • Brahamsha, B.A. (1996). Genetic manipulation system for oceanic cyanobacteria of the genus Synechococcus. Applied and Environmental Microbiology, 62: 1747–1751.
  • Breseghello, F. & Coelho, A.S.G. (2013). Traditional and modern plant breeding methods with examples in rice (Oryza sativa L.). Journal of Agricultural and Food Chemistry, 61: 8277–8286.
  • Brown, L.E., Sprecher, S.L. & Keller, L.R. (1991). Introduction of exogenous DNA into Chlamydomonas reinhardtii by electroporation. Molecular and Cellular Biology, 11: 2328–2332.
  • Carroll, D. (2011). Genome engineering with zinc-finger nucleases. Genetics, 188: 773–782.
  • Carthew, R.W. & Sontheimer, E.J. (2009). Origins and mechanisms of miRNAs and siRNAs. Cell, 136: 642–655.
  • Castel, S.E. & Martienssen, R.A. (2013). RNA interference (RNAi) in the nucleus: roles for small RNA in transcription, epigenetics and beyond. Nature Reviews Genetics, 14: 100–112.
  • Cerutti, H., Ma, X., Msanne, J. & Repas, T. (2011). RNA-mediated silencing in algae: biological roles and tools for analysis of gene function. Eukaryotic Cell, 10: 1164–1172.
  • Cha, T.S., Chen, C.F., Yee, W., Aziz, A. & Loh, S.H. (2011). Cinnamic acid, coumarin and vanillin: alternative phenolic compounds for efficient Agrobacterium-mediated transformation of the unicellular green alga, Nannochloropsis sp. Journal of Microbiological Methods, 84: 430–434.
  • Char, S.N., Neelakandan, A.K., Nahampun, H., Frame, B., Main, M., Spalding, M.H., Becraft, P.W., Meyers, B.C., Walbot, V., Wang, K. & Yang, B. (2017). An Agrobacterium-delivered CRISPR/Cas9 system for high-frequency targeted mutagenesis in maize. Plant Biotechnology Journal, 15: 257–268.
  • Charrier, B., Rolland, E., Gupta, V. & Reddy, C.R.K. (2015). Production of genetically and developmentally modified seaweeds: exploiting the potential of artificial selection techniques. Frontiers in Plant Science, 6: 127.
  • Chen, Y., Wang, Y., Sun, Y., Zhang, L & Li, W. (2001). Highly efficient expression of rabbit neutrophil peptide-1 gene in Chlorella ellipsoidea cells. Current Genetics, 39: 365–370.
  • Cheney, D., Metz, B. & Stiller, J. (2001). Agrobacterium-mediated genetic transformation in the macroscopic marine red alga Porphyra yezoensis. Journal of Phycology, 37: 11.
  • Cheng, R., Ma, R., Li, K., Rong, H., Lin, X., Wang, Z., Yang, S. & Ma, Y. (2012). Agrobacterium tumefaciens mediated transformation of marine microalgae Schizochytrium. Microbiological Research, 167: 179–186.
  • Chepurnov, V.A., Chaerle, P., Roef, L., Meirhaeghe, A. & Vanhoutte, K. (2011). Classical breeding in diatoms: scientific background and practical perspectives. In The Diatom World (Seckbach, J. & Kociolek, J.P., editors), 167–194. Springer, Dordrecht.
  • Chiang, M.S., Chong, C., Landry, B.S. & Crete, R. (1993). Cabbage Brassica oleracea subsp. capitata L. In Genetic Improvement of Vegetable Crops (Kalloo, G. & Bergh, B.O., editors), 113–155. Pergamon Press, New York.
  • Cho, S.W., Kim, S., Kim, J.M. & Kim, J.S. (2013). Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nature Biotechnology, 31: 230–232.
  • Cho, S.W., Kim, S., Kim, Y., Kweon, J., Kim, H.S., Bae, S. & Kim, J.S. (2014). Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome Research, 24: 132–141.
  • Choi, J.I., Yoon, M., Joe, M., Park, H., Lee, S.G., Han, S.J. & Lee, P.C. (2014). Development of microalga Scenedesmus dimorphus mutant with higher lipid content by radiation breeding. Bioprocess and Biosystems Engineering, 37: 2437–2444.
  • Chow, K.C. & Tung, W.L. (1999). Electrotransformation of Chlorella vulgaris. Plant Cell Reports, 18: 778–780.
  • Church, G.M., Elowitz, M.B., Smolke, C.D., Voigt, C.A. & Weiss, R. (2014). Realizing the potential of synthetic biology. Nature Reviews Molecular Cell Biology, 15: 289–294.
  • Cock, J.M. et al. (2010). The Ectocarpus genome and the independent evolution of multicellularity in brown algae. Nature, 465: 617–621.
  • 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: 819–823.
  • Cordero, B.F., Obraztsova, I., Couso, I., Leon, R., Vargas, M.A. & Rodriguez, H. (2011). Enhancement of lutein production in Chlorella sorokiniana (Chorophyta) by improvement of culture conditions and random mutagenesis. Marine Drugs, 9: 1607–1624.
  • Costa, J.A.V. & de Morais, M.G. (2013). Microalgae for food production. In Fermentation Processes Engineering in the Food Industry (Soccol, C.R., Pandey, A. & Larroche, C., editors), 405. CRC Press, Boca Raton, FL.
  • Couso, I., Cordero, B.F., Vargas, M.Á. & Rodríguez, H. (2012). Efficient heterologous transformation of Chlamydomonas reinhardtii npq2 mutant with the zeaxanthin epoxidase gene isolated and characterized from Chlorella zofingiensis. Marine Drugs, 10: 1955–1976.
  • Cradick, T.J., Qiu, P., Lee, C.M., Fine, E.J. & Bao, G. (2014). COSMID: a web-based tool for identifying and validating CRISPR/Cas off-target sites. Molecular Therapy Nucleic Acids, 3: e214.
  • Crisp, P. & Tapsell, C.R. (1993). Cauliflower Brassica oleracea L. In Genetic Improvement of Vegetable Crops (Kalloo, G. & Bergh, B.O., editors), 157–178. Pergamon Press, New York.
  • Daboussi, F., Leduc, S., Maréchal, A., Dubois, G., Guyot, V., Perez-Michaut, C., Amato, A., Falciatore, A., Juillerat, A., Beurdeley, M., Voytas, D.F., Cararec, L. & Duchateau, P. (2014). Genome engineering empowers the diatom Phaeodactylum tricornutum for biotechnology. Nature Communications, 5: 3831.
  • Dale, P.J. (1999). Public reactions and scientific responses to transgenic crops. Current Opinion in Biotechnology, 10: 203–208.
  • Dauvillée, D., Delhaye, S., Gruyer, S., Slomianny, C., Moretz, S.E., d’Hulst, C., Long, C.A., Ball, S.G. & Tomavo, S. (2010). Engineering the chloroplast targeted malarial vaccine antigens in Chlamydomonas starch granules. PLoS ONE, 5: e15424.
  • Davies, J.P., Yildiz, F. & Grossman, A.R. (1994). Mutants of Chlamydomonas with aberrant responses to sulphur deprivation. The Plant Cell, 6: 53–63.
  • Day, A. & Goldschmidt-Clermont, M. (2011). The chloroplast transformation toolbox: selectable markers and marker removal. Plant Biotechnology Journal, 9: 540–553.
  • Deng, X., Cai, J. & Fei, X. (2013). Effect of the expression and knockdown of citrate synthase gene on carbon flux during triacylglycerol biosynthesis by green algae Chlamydomonas reinhardtii. BMC Biochemistry, 14: 38.
  • De Riso, V., Raniello, R., Maumus, F., Rogato, A., Bowler, C. & Falciatore, A. (2009). Gene silencing in the marine diatom Phaeodactylum tricornutum. Nucleic Acids Research, 37: e96.
  • Dexter, J. & Fu, P. (2009). Metabolic engineering of cyanobacteria for ethanol production. Energy and Environmental Science, 2: 857–864.
  • Dianov, G.L. & Hubscher, U. (2013). Mammalian base excision repair: the forgotten archangel. Nucleic Acids Research, 41: 3483–3490.
  • Doetsch, N.A., Favreau, M.R., Kuscuoglu, N., Thompson, M.D. & Hallick, R.B. (2001). Chloroplast transformation in Euglena gracilis: splicing of a group III twintron transcribed from a transgenic psbK operon. Current Genetics, 39: 49–60.
  • Doron, L., Segal, N. & Shapira, M. (2016). Transgene expression in microalgae — from tools to applications. Frontiers in Plant Science, 7: 505.
  • Dragosits, M. & Mattanovich, D. (2013). Adaptive laboratory evolution – principles and applications for biotechnology. Microbial Cell Factories, 12: 64.
  • Dreesen, I.A.J., Hamri, G.C.E. & Fussenegger, M. (2010). Heat-stable oral alga-based vaccine protects mice from Staphylococcus aureus infection. Journal of Biotechnology, 145: 273–280.
  • Dunahay, T.G. (1993). Transformation of Chlamydomonas reinhardtii with silicon carbide whiskers. Biotechniques, 15: 452–455, 457–458, 460.
  • Dunahay, T.G., Jarvis, E.E. & Roessler, P.G. (1995). Genetic transformation of the diatoms Cyclotella cryptica and Navicula saprophila. Journal of Phycology, 31: 1004–1012.
  • Durai, S., Mani, M., Kandavelou, K., Wu, J., Porteus, M.H. & Chandrasegaran, S. (2005). Zinc finger nucleases: custom-designed molecular scissors for genome engineering of plant and mammalian cells. Nucleic Acids Research, 33: 5978–5990.
  • Eamens, A.L., McHale, M. & Waterhouse, P.M. (2014). The use of artificial microRNA technology to control gene expression in Arabidopsis thaliana. Methods in Molecular Biology, 1062: 211–224.
  • Economou, C., Wannathong, T., Szaub, J. & Purton, S. (2014). A simple, low-cost method for chloroplast transformation of the green alga Chlamydomonas reinhardtii. Methods in Molecular Biology, 1132: 401–411.
  • Falciatore, A., Casotti, R., Leblanc, C., Abrescia, C. & Bowler, C. (1999). Transformation of nonselectable reporter genes in marine diatoms. Marine Biotechnology, 1: 239–251.
  • Feng, S., Feng, W., Zhao, L., Gu, H., Li, Q., Shi, K., Guo, S. & Zhang, N. (2014). Preparation of transgenic Dunaliella salina for immunization against white spot syndrome virus in crayfish. Archives of Virology, 159: 519–525.
  • Feng, S.Y., Xue, L.X., Liu, H.T. & Lu, P.J. (2009). Improvement of efficiency of genetic transformation for Dunaliella salina by glass beads method. Molecular Biology Reports, 36: 1433–1439.
  • Fire, A., Xu, S., Montgomery, M.K., Kostas, S.A., Driver, S.E. & Mello, C.C. (1998). Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature, 391: 806–811.
  • Franklin, S., Ngo, B., Efuet, E. & Mayfield, S.P. (2002). Development of a GFP reporter gene for Chlamydomonas reinhardtii chloroplast. The Plant Journal, 30: 733–744.
  • Fu, Y., Foden, J.A., Khayter, C., Maeder, M.L., Reyon, D., Joung, J.K. & Sander, J.D. (2013). High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nature Biotechnology, 31: 822–826.
  • Fu, W., Guðmundsson, O., Paglia, G., Herjólfsson, G., Andrésson, O.S., Palsson, B.O. & Brynjólfsson, S. (2013). Enhancement of carotenoid biosynthesis in the green microalga Dunaliella salina with light-emitting diodes and adaptive laboratory evolution. Applied Microbiology and Biotechnology, 97: 2395–2403.
  • Fu, W., Chaiboonchoe, A., Khraiwesh, B., Nelson, D.R., Al-Khairy, D., Mystikou, A., Alzahmi, A. & Salehi-Ashtiani, K. (2016). Algal cell factories: approaches, applications, and potentials. Marine Drugs, 14: 225.
  • Fujita, Y. & Saito, M. (1990). Protoplast isolation and fusion in Porphyra (Bangiales, Rhodophyta). Developments in Hydrobiology, 58: 161–166.
  • Fukuda, S., Mikami, K., Uji, T., Park, E.J., Ohba, T., Asada, K., Kitade, Y., Endo, H., Kato, I. & Saga, N. (2008). Factors influencing efficiency of transient gene expression in the red macrophyte Porphyra yezoensis. Plant Science, 174: 329–339.
  • Gan, S.Y., Qin, S., Othman, R.Y., Yu, D. & Phang, S.M. (2003). Transient expression of lacZ in particle bombarded Gracilaria changii (Gracilariales, Rhodophyta). Journal of Applied Phycology, 15: 345–349.
  • Gan, S.Y., Lim, P.E. & Phang, S.M. (2016). Genetic and metabolic engineering of microalgae. In Algae Biotechnology, Green Energy and Technology (Bux, F. & Chisti, Y., editors), 317–344. Springer International, Cham.
  • 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: 2579–2586.
  • Gay, P.B. & Gillespie, S.H. (2005). Antibiotic resistance markers in genetically modified plants: a risk to human health? Lancet Infectious Diseases, 5: 637–646.
  • Geng, D., Wang, Y., Wang, P. Li, W. & Sun, Y. (2003). Stable expression of hepatitis B surface antigen gene in Dunaliella salina (Chlorophyta). Journal of Applied Phycology, 15: 451–456.
  • Georgianna, D.R. & Mayfield, S.P. (2012). Exploiting diversity and synthetic biology for the production of algal biofuels. Nature, 488: 329–335.
  • Ghildiyal, M. & Zamore, P. D. (2009). Small silencing RNAs: an expanding universe. Nature Reviews Genetics, 10: 94–108.
  • Glass, D.J. (2015). Government regulation of the uses of genetically modified algae and other microorganisms in biofuel and bio-based chemical production. In Algal Biorefineries (Prokop, A., Bajpai, R.K. & Zappi, M.E., editors), 23–60. Springer International Publishing, Cham.
  • Gong, Q., Yu, W., Dai, J., Liu, H., Xu, R., Guan, H. & Pan, K. (2007). Efficient gusA transient expression in Porphyra yezoensis protoplasts mediated by endogenous beta-tubulin flanking sequences. Journal of Ocean University of China, 6: 21–25.
  • Grau, J., Boch, J. & Posch, S. (2013). TALENoffer: genome-wide TALEN off-target prediction. Bioinformatics, 29: 2931–2932.
  • Gressel, J. (1999). Tandem constructs: preventing the rise of superweeds. Trends in Biotechnology, 17: 361–366.
  • Gressel, J., van der Vlugt, C.J.B. & Bergmans, E.N. (2013). Environmental risks of large scale cultivation of microalgae: mitigation of spills. Algal Research, 2: 286–298.
  • Gressel, J., van der Vlugt, C.J.B. & Bergmans, E.N. (2014). Cultivated microalgae spills: hard to predict/easier to mitigate risks. Trends in Biotechnology, 32: 65–69.
  • Guilinger, J.P., Thompson, D.B. & Liu, D.R. (2014). Fusion of catalytically inactive Cas9 to FokI nuclease improves the specificity of genome modification. Nature Biotechnology, 32: 577–582.
  • Guo, S., Zhao, X., Tang, Y., Wan, C., Alam, M.A., Ho, S., Bai, F. & Chang, J. (2013). Establishment of an efficient genetic transformation system in Scenedesmus obliquus. Journal of Biotechnology, 163: 61–68.
  • Gupta, V., Kumari, P. & Reddy, C. (2015). Development and characterization of somatic hybrids of Ulva reticulata Forsskål (×) Monostroma oxyspermum (Kutz.) Doty. Frontiers in Plant Science, 6: 3.
  • Hado, M., Okauchhi, M., Murase, N. & Mizukami, Y. (2003). Transient expression of GUS gene using Rubisco gene promoter in the protoplasts of Porphyra yezoensis. Aquaculture Science, 51: 355–360.
  • Hallmann, A. (2007). Algal transgenics and biotechnology. Transgenic Plant Journal, 1: 81–98.
  • Hallmann, A., Rappel, A. & Sumper, M. (1997). Gene replacement by homologous recombination in the multicellular green alga Volvox carteri. Proceedings of the National Academy of Sciences of the United States of America, 94: 7469–7474.
  • Harper, M. & Lee, C.J. (2012). Genome-wide analysis of mutagenesis bias and context sensitivity of N-methyl-N’-nitro-N-nitrosoguanidine (NTG). Mutation Research, 731: 64–67.
  • Hawkins, R.L. & Nakamura, M. (1999). Expression of human growth hormone by the eukaryotic alga, Chlorella. Current Microbiology, 38: 335–341.
  • He, P., Yao, Q., Chen, Q., Guo, M., Xiong, A., Wu, W. & Ma, J. (2001). Transferring and expression of glucose oxidase gene in Porphyra yezoensis. Journal of Phycology, 37: 23.
  • Heigwer, F., Kerr, G., Walther, N., Glaeser, K., Pelz, O., Breinig, M. & Boutros, M. (2013). E-TALEN: a web tool to design TALENs for genome engineering. Nucleic Acids Research, 41: e190.
  • Henley, W.J., Litaker, R.W., Novoveská, L., Duke, C.S., Quemada, H.D. & Sayre, R.T. (2013). Initial risk assessment of genetically modified (GM) microalgae for commodity-scale biofuel cultivation. Algal Research, 2: 66–77.
  • Hirata, R., Takahashi, M., Saga, N. & Mikami, K. (2011). Transient gene expression system established in Porphyra yezoensis is widely applicable in Bangiophycean algae. Marine Biotechnology, 13: 1038–1047.
  • Hopes, A., Nekrasov, V., Kamoun, S. & Mock, T. (2016). Editing of the urease gene by CRISPR-Cas in the diatom Thalassiosira pseudonana. Plant Methods, 12: 49.
  • Hsu, P.D., Scott, D.A., Weinstein, J.A., Ran, F.A., Konermann, S., Agarwala, V., Li, Y., Fine, E.J., Wu, X., Shalem, O., Cradick, T.J., Marraffini, L.A., Bao, G. & Zhang, F. (2013). DNA targeting specificity of RNA-guided Cas9 nucleases. Nature Biotechnology, 31: 827–832.
  • Huang, A., He, L. & Wang, G. (2011). Identification and characterization of microRNAs from Phaeodactylum tricornutum by high-throughput sequencing and bioinformatics analysis. BMC Genomics, 12: 337.
  • Huang, X., Weber, J.C., Hinson, T.K., Mathieson, A.C. & Minocha, S.C. (1996). Transient expression of the GUS reporter gene in the protoplasts and partially digested cells of Ulva lactuca L (Chlorophyta). Botanica Marina, 39: 467–474.
  • Huddy, S.M., Meyers, A.E. & Coyne, V.E. (2012). Transformation of lacZ using different promoters in the commercially important red alga, Gracilaria gracilis. African Journal of Biotechnology, 11: 1879–1885.
  • Hutchinson, G.E. (1961). The paradox of the plankton. American Naturalist, 95: 137–145.
  • Iseki, M., Matsunaga, S., Murakami, A., Ohno, K., Shiga, K., Yoshida, K., Sugai, M., Takahashi, T., Hori, T. & Watanabe, M. (2002). A blue-light-activated adenylyl cyclase mediates photoavoidance in Euglena gracilis. Nature, 415: 1047–1051.
  • Ishikawa, T., Nishikawa, H., Gao, Y., Sawa, Y., Shibata, H., Yabuta, Y., Maruta, T. & Shigeoka, S. (2008). The pathway via D-galacturonate/L-galactonate is significant for ascorbate biosynthesis in Euglena gracilis: identification and functional characterization of aldonolactonase. Journal of Biological Chemistry, 283: 31133–31141.
  • Jacobs, T.B., LaFayette, P.R., Schmitz, R.J. & Parrott, W.A. (2015). Targeted genome modifications in soybean with CRISPR/Cas9. BMC Biotechnology, 15: 16.
  • Jakobiak, T., Mages, W., Scharf, B., Babinger, P., Stark, K. & Schmitt, R. (2004). The bacterial paromomycin resistance gene, aphH, as a dominant selectable marker in Volvox carteri. Protist, 155: 381–393.
  • Jia, Y., Xue, L., Liu, H. & Li, J. (2009). Characterization of the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) gene from the halotolerant alga Dunaliella salina and inhibition of its expression by RNAi. Current Microbiology, 58: 426–431.
  • Jiang, G.Z., Lu, Y.M., Niu, X.L. & Xue, L.X. (2005). The actin gene promoter-derived bar as dominant selectable marker for nuclear transformation of Dunaliella salina. Yi Chuan Xue Bao, 32: 424–433.
  • Jiang, P., Qin, S. & Tseng, C.K. (2002). Expression of hepatitis B surface antigen gene (HBsAg) in Laminaria japonica (Laminariales, Phaeophyta). Chinese Science Bulletin, 47: 1438–1440.
  • Jiang, P., Qin, S. & Tseng, C.K. (2003). Expression of the lacZ reporter gene in sporophytes of the seaweed Laminaria japonica (Phaeophyceae) by gametophyte-targeted transformation. Plant Cell Reports, 21: 1211–1216.
  • Jiang, W., Brueggeman, A.J., Horken, K.M., Plucinak, T.M. & Weeks, D.P. (2014). Successful transient expression of Cas9 and single guide RNA genes in Chlamydomonas reinhardtii. Eukaryotic Cell, 13: 1465–1469.
  • Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., Doudna, J.A. & Charpentier, E. (2012). A programmable dual-RNA–guided DNA endonuclease in adaptive bacterial immunity. Science, 337: 816–821.
  • Jinkerson, R.E. & Jonikas, M.C. (2015). Molecular techniques to interrogate and edit the Chlamydomonas nuclear genome. Plant Journal, 82: 393–412.
  • Joung, J.K. & Sander, J.D. (2013). TALENs: a widely applicable technology for targeted genome editing. Nature Reviews Molecular Cell Biology, 14: 49–55.
  • Kakinuma, M., Ikeda, M., Coury, D.A., Tominaga, H., Kobayashi, I. & Amano, H. (2009). Isolation and characterization of the rbcS genes from a sterile mutant of Ulva pertusa (Ulvales, Chlorophytea) and transient gene expression using the rbcS gene promoter. Fisheries Science, 75: 1015–1028.
  • Kasai, Y., Oshima, K., Ikeda, F., Abe, J., Yoshimitsu, Y. & Harayama, S. (2015). Construction of a self-cloning system in the unicellular green alga Pseudochoricystis ellipsoidea. Biotechnology for Biofuels, 8: 94.
  • Kathiresan, S., Chandrashekar, A., Ravishankar, G.A. & Sarada, R. (2009). Agrobacterium-mediated transformation in the green alga Haematococcus pluvialis (Chlorophyceae, Volvocales). Journal of Phycology, 45: 642–649.
  • Ketting, R.F. (2011). The many faces of RNAi. Developmental Cell, 20: 148–161.
  • Key, S., Ma, J.K.-C. & Drake, P.M. (2008). Genetically modified plants and human health. Journal of the Royal Society of Medicine, 101: 290–298.
  • Khawaja, I.B., Kim, M., Stahl, U. & Cho, M. (2016). Microalgae engineering toolbox: selectable and screenable markers. Biotechnology and Bioprocess Engineering, 21: 224–235.
  • Kilian, O., Benemann, C.S.E., Niyogi, K.K. & Vick, B. (2011). High-efficiency homologous recombination in the oil-producing alga Nannochloropsis sp. Proceedings of the National Academy of Sciences of the United States of America, 108: 21265–21269.
  • Kim, D., Kim, Y.T., Cho, J.J., Bae, J.H., Hur, S.B., Hwang, I. & Choi, T.J. (2002). Stable integration and functional expression of flounder growth hormone gene in transformed microalgae Chlorella ellipsoidea. Marine Biotechnology, 4: 63–73.
  • 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: 1012–1019.
  • Kim, S., Lee, Y.C., Cho, D.H., Lee, H.U., Huh, Y.S., Kim, G.J. & Kim, H.S. (2014). A simple and non-invasive method for nuclear transformation of intact-walled Chlamydomonas reinhardtii. PLoS ONE, 9: e101018.
  • Kindle, K.L. (1990). High-frequency nuclear transformation of Chlamydomonas reinhardtii. Proceedings of the National Academy of Sciences of the United States of America, 87: 1228–1232.
  • Kito, H., Kunimoto, M., Kamanishi, Y. & Mizukami, Y. (1998). Protoplast fusion between Monostroma nitidum and Porphyra yezoensis and subsequent growth of hybrid plants. Journal of Applied Phycology, 10: 15–21.
  • Koblenz, B. & Lechtreck, K.F. (2005). The NIT1 promoter allows inducible and reversible silencing of centrin in Chlamydomonas reinhardtii. Eukaryotic Cell, 4: 1959–1962.
  • Kong, F., Zhao, H., Liu, W., Li, N. & Mao, Y. (2017). Construction of plastid expression vector and development of genetic transformation system for the seaweed Pyropia yezoensis. Marine Biotechnology, 19: 147–156.
  • Kuang, M., Wang, S.J., Li, Y., Shen, D.L. & Zeng, C.K. (1998). Transient expression of exogenous GUS gene in Porphyra yezoensis (Rhodophyta). Chinese Journal of Oceanology and Limnology, 16: 56–61.
  • Kübler, J.E., Minocha, S.C. & Mathieson, A.C. (1994). Transient expression of the GUS reporter gene in protoplasts of Porphyra miniata (Rhodophyta). Journal of Marine Biotechnology, 1: 165–169.
  • Kulkarni, M.M., Booker, M., Silver, S.J., Friedman, A., Hong, P., Perrimon, N. & Mathey-Prevot, B. (2006). Evidence of off-target effects associated with long dsRNAs in Drosophila melanogaster cell-based assays. Nature Methods, 3: 833–838.
  • Kumar, S.V., Misquitta, R.W., Reddy, V.S., Rao, B.J. & Rajam, M.V. (2004). Genetic transformation of the green alga – Chlamydomonas reinhardtii by Agrobacterium tumefaciens. Plant Science, 166: 731–738.
  • Kurtzman, A.M. & Cheney, D.P. (1991). Direct gene transfer and transient expression in a marine red alga using the biolistic method. Journal of Phycology, 27(Supplement): 42.
  • Lapidot, M., Raveh, D., Sivan, A., Arad, S.M. & Shapira, M. (2002). Stable chloroplast transformation of the unicellular red alga Porphyridium species. Plant Physiology, 129: 7–12.
  • Ledford, H. (2016). CRISPR: gene editing is just the beginning. Nature, 531: 156–159.
  • Lee, B., Choi, G.G., Choi, Y.E., Sung, M., Park, M.S. & Yang, J.W. (2014). Enhancement of lipid productivity by ethyl methane sulfonate-mediated random mutagenesis and proteomic analysis in Chlamydomonas reinhardtii. Korean Journal of Chemical Engineering, 31: 1036–1042.
  • Lei, Y., Lu, L., Liu, H.Y., Li, S., Xing, F. & Chen, L.L. (2014). CRISPR-P: a web tool for synthetic single-guide RNA design of CRISPR-system in plants. Molecular Plant, 7: 1494–1496.
  • León, R., Couso, I. & Fernández, E. (2007). Metabolic engineering of ketocarotenoids biosynthesis in the unicelullar microalga Chlamydomonas reinhardtii. Journal of Biotechnology, 130: 143–152.
  • Lerche, K. & Hallmann A. (2009). Stable nuclear transformation of Gonium pectorale. BMC Biotechnology, 9: 64.
  • Levitan, O., Dinamarca, J., Zelzion, E., Lun, D.S., Guerra, L.T., Kim, M.K., Kim, J., Van Mooy, B.A.S., Bhattacharya, D. & Falkowski, P.G. (2014). Remodeling of intermediate metabolism in the diatom Phaeodactylum tricornutum under nitrogen stress. Proceedings of the National Academy of Sciences of the United States of America, 112: 412–417.
  • Li, D., Wang, L., Zhao, Q., Wei, W. & Sun, Y. (2015). Improving high carbon dioxide tolerance and carbon dioxide fixation capability of Chlorella sp. by adaptive laboratory evolution. Bioresource Technology, 185: 269–275.
  • Li, F., Qin, S., Jiang, P., Wu, Y. & Zhang, W. (2009). The integrative expression of GUS gene driven by FCP promoter in the seaweed Laminaria japonica (Phaeophyta). Journal of Applied Phycology, 21: 287–293.
  • Li, J.F., Norville, J.E., Aach, J., McCormack, M., Zhang, D., Bush, J., Church, G.M. & Sheen, J. (2013). Multiplex and homologous recombination-mediated genome editing in Arabidopsis and Nicotiana benthamiana using guide RNA and Cas9. Nature Biotechnology, 31: 688–691.
  • Liang, C., Zhang, X., Zou, J., Xu, D., Su, F. & Ye, N. (2010). Identification of miRNA from Porphyra yezoensis by high-throughput sequencing and bioinformatics analysis. PLoS ONE, 5: e10698.
  • Liang, S., Guo, L., Lin, G., Zhang, Z., Ding, H., Wang, Y. & Yang, G. (2017). Improvement of Nannochloropsis oceanica growth performance through chemical mutation and characterization of fast growth physiology by transcriptome profiling. Chinese Journal of Oceanology and Limnology, 35: 792–802.
  • Lin, H., & Qin, S. (2014). Tipping points in seaweed genetic engineering: scaling up opportunities in the next decade. Marine Drugs, 12: 3025–3045.
  • Liu, H., Wei, Z., Dominguez, A., Li, Y., Wang, X. & Qi, L.S. (2015). CRISPR-ERA: a comprehensive design tool for CRISPR-mediated gene editing, repression and activation. Bioinformatics, 31: 3676–3678.
  • Liu, H.Q., Yu, W.G., Dai, J.X., Gong, Q.H., Yang, K.F. & Zhang, Y.P. (2003). Increasing the transient expression of GUS gene in Porphyra yezoensis by 18S rDNA targeted homologous recombination. Journal of Applied Phycology, 15: 371–377.
  • Liu, J., Sun, Z., Gerken, H., Huang, J., Jiang, Y. & Chen, F. (2014). Genetic engineering of the green alga Chlorella zofingiensis: a modified norflurazon-resistant phytoene desaturase gene as a dominant selectable marker. Applied Microbiology and Biotechnology, 98: 5069–5079.
  • Liu, L., Wang, Y., Zhang, Y., Chen, X., Zhang, P. & Ma, S. (2013). Development of a new method for genetic transformation of the green alga Chlorella ellipsoidea. Molecular Biotechnology, 54: 211–219.
  • Liu, S., Zhao, Y., Liu, L., Ao, X., Ma, L., Wu, M. & Ma, F. (2015). Improving cell growth and lipid accumulation in green microalgae Chlorella sp. via UV irradiation. Applied Biochemistry and Biotechnology, 175: 3507–3518.
  • Lozano, J.C., Schatt, P., Botebol, H., Verge, V., Lesuisse, E., Blain, S., Carre, I.A. & Bouget, F.Y. (2014). Efficient gene targeting and removal of foreign DNA by homologous recombination in the picoeukaryote Ostreococcus. Plant Journal, 78: 1073–1083.
  • Lumbreras, V., Stevens, D.R. & Purton, S. (1998). Efficient foreign gene expression in Chlamydomonas reinhardtii mediated by an endogenous intron. The Plant Journal, 14: 441–447.
  • Maghari, B.M. & Ardekani, A.M. (2011). Genetically modified foods and social concerns. Avicenna Journal of Medical Biotechnology, 3: 109–117.
  • Manuell, A.L., Beligni, M.V., Elder, J.H., Siefker, D.T., Tran, M., Weber, A., McDonald, T.L. & Mayfield, S.P. (2007). Robust expression of a bioactive mammalian protein in Chlamydomonas chloroplast. Plant Biotechnology Journal, 5: 402–412.
  • Maruyama, M., Horáková, I., Honda, H., Xing, X., Shiragami, N. & Unno, H. (1994). Introduction of foreign DNA into Chlorella saccharophila by electroporation. Biotechnology Techniques, 8: 821–826.
  • Matsunaga, T., Takeyama, H. & Nakamura, N. (1990). Characterization of cryptic plasmids from marine cyanobacteria and construction of a hybrid plasmid potentially capable of transformation of marine cyanobacterium, Synechococcus sp. and its transformation. Applied Biochemistry and Biotechnology, 24/25: 151–160.
  • Mayfield, S.P., Franklin, S.E. & Lerner, R.A. (2003). Expression and assembly of a fully active antibody in algae. Proceedings of the National Academy of Sciences of the United States of America, 100: 438–442.
  • Mayfield, S.P., Manuell, A.L., Chen, S., Wu, J., Tran, M., Siefker, D., Muto, M. & Marin-Navarro, J. (2007). Chlamydomonas reinhardtii chloroplasts as protein factories. Current Opinion in Biotechnology, 18: 126–133.
  • Meneses, I. & Santelices, B. (1999). Strain selection and genetic variation in Gracilaria chilensis (Gracilariales, Rhodophyta). Journal of Applied Phycology, 11: 241–246.
  • Mikami, K. (2014). A technical breakthrough close at hand: feasible approaches toward establishing a gene-targeting genetic transformation system in seaweeds. Frontiers in Plant Science, 5: 498.
  • Mikami, K., Uji, T., Li, L., Takahashi, M., Yasui, H. & Saga, N. (2009). Visualization of phosphoinositides via the development of the transient expression system of a cyan fluorescent protein in the red alga Porphyra yezoensis. Marine Biotechnology, 11: 563–569.
  • Miyagawa, A., Okami, T., Kira, N., Yamaguchi, H., Ohnishi, K. & Adachi, M. (2009). Research note: high efficiency transformation of the diatom Phaeodactylum tricornutum with a promoter from the diatom Cylindrotheca fusiformis. Phycological Research, 57: 142–146.
  • Miyagawa, A., Okami, T., Kira, N., Yamaguchi, H., Ohnishi, K. & Adachi, M. (2011). Stable nuclear transformation of the diatom Chaetoceros sp. Phycological Research, 59: 113–119.
  • Miyaoka, Y., Berman, J.R., Cooper, S.B., Mayerl, S.J., Chan, A.H., Zhang, B., Karlin-Neumann, G.A. & Conklin, B.R. (2016). Systematic quantification of HDR and NHEJ reveals effects of locus, nuclease, and cell type on genome-editing. Scientific Reports, 6: 23549.
  • Mizukami, Y., Okauchi, M., Kito, H., Ishimoto, S., Ishida, T. & Fuseya, M. (1995). Culture and development of electrically fused protoplasts from red marine algae, Porphyra yezoensis and P. suborbiculata. Aquaculture, 132: 361–367.
  • Mizukami, Y., Hado, M., Kito, H., Kunimoto, M. & Murase, N. (2004). Reporter gene introduction and transient expression in protoplasts of Porphyra yezoensis. Journal of Applied Phycology, 16: 23–29.
  • Molnár, A., Schwach, F., Studholme, D.J., Thuenemann, E.C. & Baulcombe, D.C. (2007). miRNAs control gene expression in the single-cell alga Chlamydomonas reinhardtii. Nature, 447: 1126–1129.
  • Molnár, A., Bassett, A., Thuenemann, E., Schwach, F., Karkare, S., Ossowski, S., Weigel, D. & Baulcombe, D. (2009). Highly specific gene silencing by artificial microRNAs in the unicellular alga Chlamydomonas reinhardtii. The Plant Journal, 58: 165–174.
  • Muto, M., Henry, R.E. & Mayfield, S.P. (2009). Accumulation and processing of a recombinant protein designed as a cleavable fusion to the endogenous Rubisco LSU protein in Chlamydomonas chloroplast. BMC Biotechnology, 9: 26.
  • Neupert, J., Shao, N., Lu, Y. & Bock, R. (2012). Genetic transformation of the model green alga Chlamydomonas reinhardtii. Methods in Molecular Biology, 847: 35–47.
  • Nickelsen, J. (1999). Transcripts containing the 5’ untranslated regions of the plastid genes psbA and psbB from higher plants are unstable in Chlamydomonas reinhardtii chloroplasts. Molecular and General Genetics, 262: 768–771.
  • Niu, Y.F., Zhang, M.H., Xie, W.H., Li, J.N., Gao, Y.F., Yang, W.D., Liu, J.S. & Li, H.Y. (2011). A new inducible expression system in a transformed green alga, Chlorella vulgaris. Genetics and Molecular Research, 10: 3427–3434.
  • Nymark, M., Sharma, A.K., Sparstad, T., Bones, A.M. & Winge, P. (2016). A CRISPR/Cas9 system adapted for gene editing in marine algae. Scientific Reports, 6: 24951.
  • Ohnuma, M., Yokoyama, T., Inouye, T., Sekine, Y. & Tanaka, K. (2008). Polyethylene glycol (PEG)-mediated transient gene expression in a red alga, Cyanidioschyzon merolae 10D. Plant and Cell Physiology, 49: 117–120.
  • Ohresser, M., Matagne, R.F. & Loppes, R. (1997). Expression of the arylsulphatase reporter gene under the control of the nit1 promoter in Chlamydomonas reinhardtii. Current Genetics, 31: 264–271.
  • Ortiz-Marquez, J.C.F., Do Nascimento, M., Zehr, J.P. & Curatti, L. (2013). Genetic engineering of multispecies microbial cell factories as an alternative for bioenergy production. Trends in Biotechnology, 31: 521–529.
  • Pabo, C.O., Peisach, E. & Grant, R.A. (2001). Design and selection of novel Cys2His2 zinc finger proteins. Annual Reviews of Biochemistry, 70: 313–340.
  • Pan, Y., Shen, N., Jung-Klawitter, S., Betzen, C., Hoffmann, G.F., Hoheisel, J.D. & Blau, N. (2016). CRISPR RNA-guided FokI nucleases repair a PAH variant in a phenylketonuria model. Scientific Reports, 6: 35794.
  • Patra, K.P., Li, F., Carter, D., Gregory, J.A., Baga, S., Reed, S.G., Mayfield, S.P. & Vinetz, J.M. (2015). Alga-produced malaria transmission-blocking vaccine candidate Pfs25 formulated with a human use-compatible potent adjuvant induces high-affinity antibodies that block Plasmodium falciparum infection of mosquitoes. Infection and Immunity, 83: 1799–1808.
  • Patwary, M.U. & van der Meer, J.P. (1992). Genetics and breeding of cultivated seaweeds. Algae, 7: 281–318.
  • Perrineau, M.M., Gross, J., Zelzion, E., Price, D.C., Levitan, O., Boyd, J. & Bhattacharya, D. (2014). Using natural selection to explore the adaptive potential of Chlamydomonas reinhardtii. PLoS ONE, 9: e92533.
  • Poulsen, N. & Kröger, N. (2005). A new molecular tool for transgenic diatoms – control of mRNA and protein biosynthesis by an inducible promoter-terminator cassette. FEBS Journal, 272: 3413–3423.
  • Poulsen, N., Chesley, P.M. & Kroger, N. (2006). Molecular genetic manipulation of the diatom Thalassiosira pseudonana (Bacillariophyceae). Journal of Phycology, 42: 1059–1065.
  • Pratheesh, P.T., Vineetha, M. & Kurup, G.M. (2014). An efficient protocol for the Agrobacterium-mediated genetic transformation of microalga Chlamydomonas reinhardtii. Molecular Biotechnology, 56: 507–515.
  • Prieto, R., Dubus, A., Galván, A. & Fernáandez, E. (1996). Isolation and characterization of two new negative regulatory mutants for nitrate assimilation in Chlamydomonas reinhardtii obtained by insertional mutagenesis. Molecular and General Genetics, 251: 461–471.
  • Pulz, O. (2001). Photobioreactors: production systems for phototrophic microorganisms. Applied Microbiology Biotechnology, 57: 287–293.
  • Purton, S. & Rochaix, J.D. (1995). Characterisation of the ARG7 gene of Chlamydomonas reinhardtii and its application to nuclear transformation. European Journal of Phycology, 30: 141–148.
  • Qin, S., Jiang, P. & Tseng, C. (2005). Transforming kelp into a marine bioreactor. Trends in Biotechnology, 23: 264–268.
  • Qin, S., Lin, H. & Jiang, P. (2012). Advances in genetic engineering of marine algae. Biotechnology Advances, 30: 1602–1613.
  • Radakovits, R., Jinkerson, R.E., Darzins, A. & Posewitz, M.C. (2010). Genetic engineering of algae for enhanced biofuel production. Eukaryotic Cell, 9: 486–501.
  • Radakovits, R., Eduafo, P.M. & Posewitz, M.C. (2011). Genetic engineering of fatty acid chain length in Phaeodactylum tricornutum. Metabolic Engineering, 13: 89–95.
  • Ramakrishna, S., Kwaku Dad, A.B., Beloor, J., Gopalappa, R., Lee, S.K. & Kim, H. (2014). Gene disruption by cell-penetrating peptide-mediated delivery of Cas9 protein and guide RNA. Genome Research, 24: 1020–1027.
  • Ramessar, K., Peremarti, A., Gómez-Galera, S., Naqvi, S., Moralejo, M., Muñoz, P., Capell, T. & Christou, P. (2007). Biosafety and risk assessment framework for selectable marker genes in transgenic crop plants: a case of the science not supporting the politics. Transgenic Research, 16: 261–280.
  • Ran, F.A., Hsu, P.D., Lin, C.Y., Gootenberg, J.S., Konermann, S., Trevino, A.E., Scott, D.A., Inoue, A., Matoba, S., Zhang, Y. & Zhang, F. (2013). Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell, 154: 1380–1389.
  • Rasala, B.A., Muto, M., Lee, P.A., Jager, M., Cardoso, R.M.F., Behnke, C.A., Kirk, P., Hokanson, C.A., Crea, R., Mendez, M. & Mayfield, S.P. (2010). Production of therapeutic proteins in algae, analysis of expression of seven human proteins in the chloroplast of Chlamydomonas reinhardtii. Plant Biotechnology Journal, 8: 719–733.
  • Sakaguchi, T., Nakajima, K. & Matsuda, Y. (2011). Identification of the UMP synthase gene by establishment of uracil auxotrophic mutants and the phenotypic complementation system in the marine diatom Phaeodactylum tricornutum. Plant Physiology, 156: 78–89.
  • Sakuma, T., Nishikawa, A., Kume, S., Chayama, K. & Yamamoto, T. (2014). Multiplex genome engineering in human cells using all-in-one CRISPR/Cas9 vector system. Scientific Reports, 4: 5400.
  • Sandesh Kamath, B., Vidhyavathi, R., Sarada, R. & Ravishankar, G.A. (2008). Enhancement of carotenoids by mutation and stress induced carotenogenic genes in Haematococcus pluvialis mutants. Bioresource Technology, 99: 8667–8673.
  • Scaife, M.A. & Smith, A.G. (2016). Towards developing algal synthetic biology. Biochemical Society Transactions, 44: 716–722.
  • Schmollinger, S., Strenkert, D. & Schroda, M. (2010). An inducible artificial microRNA system for Chlamydomonas reinhardtii confirms a key role for heat shock factor 1 in regulating thermotolerance. Current Genetics, 56: 383–389.
  • Schroda, M., Vallon, O., Wollman, F.A. & Beck, C.F. (1999). A chloroplast-targeted heat shock protein 70 (HSP70) contributes to the photoprotection and repair of photosystem II during and after photoinhibition. Plant Cell, 11: 1165–1178.
  • Scowcroft, W.R. & Larkin, P.J. (1988). Somaclonal variation. In Applications of Plant Cell and Tissue Culture (Bock, G. & Marsh, J., editors), 21–35. John Wiley and Sons, Chichester.
  • Sharma, S.K., Nelson, D.R., Abdrabu, R., Khraiwesh, B., Jijakli, K., Arnoux, M., O’Connor, M.J., Bahmani, T., Cai, H., Khapli, S., Jagannathan, R. & Salehi-Ashtiani, K. (2015). An integrative Raman microscopy-based workflow for rapid in situ analysis of microalgal lipid bodies. Biotechnology for Biofuels, 8: 164.
  • Shimogawara, K., Fujiwara, S., Grossman, A. & Usuda, H. (1998). High-efficiency transformation of Chlamydomonas reinhardtii by electroporation. Genetics, 148: 1821–1828.
  • Shin, S.E., Lim, J.M., Koh, H.G., Kim, E.K., Kang, N.K., Jeon, S., Kwon, S., Shin, W. S., Lee, B., Hwangbo, K., Kim, J., Ye, S.H., Yun, J.Y., Seo, H., Oh, H.M., Kim, K.J., Kim, J.S., Jeong, W.J., Chang, Y.K. & Jeong, B. (2016). CRISPR/Cas9-induced knockout and knock-in mutations in Chlamydomonas reinhardtii. Scientific Reports, 6: 27810.
  • Sizova, I., Fuhrmann, M. & Hegemann, P. (2011). A Streptomyces rimosus aphVIII gene coding for a new type phosphotransferase provides stable antibiotic resistance to Chlamydomonas reinhardtii. Gene, 277: 221–229.
  • Sizova, I., Greiner, A., Awasthi, M., Kateriya, S. & Hegemann, P. (2013). Nuclear gene targeting in Chlamydomonas using engineered zinc-finger nucleases. Plant Journal, 73: 873–882.
  • Sode, K., Tatara, M., Takeyama, H., Burgess, J.G. & Matsunaga, T. (1992). Conjugative gene-transfer in marine cyanobacteria – Synechococcus sp., Synechocystis sp. and Pseudanabaena sp. Applied Microbiology and Biotechnology, 37: 369–373.
  • Sodeinde, O.A. & Kindle, K.L. (1993). Homologous recombination in the nuclear genome of Chlamydomonas reinhardtii. Proceedings of the National Academy of Sciences of the United States of America, 90: 9199–9203.
  • Son, S.H., Ahn, J.W., Uji, T., Choi, D.W., Park, E.J., Hwang, M.S., Liu, J.R., Choi, D., Mikami, K. & Jeong, W.J. (2012). Development of an expression system using the heat shock protein 70 promoter in the red macroalga, Porphyra tenera. Journal of Applied Phycology, 24: 79–87.
  • Srinivasan, R. & Gothandam, K.M. (2016). Synergistic action of D-glucose and acetosyringone on Agrobacterium strains for efficient Dunaliella transformation. PLoS ONE, 11: e0158322.
  • Steinbrenner, J. & Sandmann, G. (2006). Transformation of the green alga Haematococcus pluvialis with a phytoene desaturase for accelerated astaxanthin biosynthesis. Applied and Environmental Microbiology, 72: 7477–7484.
  • Sternberg, S.H., Redding, S., Jinek, M., Greene, E.C. & Doudna, J.A. (2014). DNA interrogation by the CRISPR RNA-guided endonuclease Cas9. Nature, 507: 62–67.
  • Sun, M., Qian, K., Su, N., Chang, H., Liu, J. & Shen, G. (2003). Foot-and-mouth disease virus VP1 protein fused with cholera toxin B subunit expressed in Chlamydomonas reinhardtii chloroplast. Biotechnology Letters, 25: 1087–1092.
  • Sun, G.H., Zhang, X.C., Sui Z.H. & Mao, Y.X. (2008). Inhibition of pds gene expression via the RNA interference approach in Dunaliella salina (Chlorophyta). Marine Biotechnology, 10: 219–226.
  • Surzycki, R., Greenham, K., Kitayama, K., Dibal, F., Wagner, R., Rochaix, J.D., Ajam, T. & Surzycki, S. (2009). Factors effecting expression of vaccines in microalgae. Biologicals, 37: 133–138.
  • Szyjka, S.J., Mandal, S., Schoepp, N.G., Tyler, B.M., Yohn, C.B., Poon, Y.S., Villareal, S., Burkart, M.D., Shurin, J.B. & Mayfield, S.P. (2017). Evaluation of phenotype stability and ecological risk of a genetically engineered alga in open pond production. Algal Research, 24: 378–386.
  • Takahashi, M., Mikami, K., Mizuta, H. & Saga, N. (2011). Identification and efficient utilization of antibiotics for the development of a stable transformation system in Porphyra yezoensis (Bangiales, Rhodophyta). Journal of Aquaculture Research and Development, S3: 002.
  • Tan, C., Qin, S., Zhang, Q. Jiang, P. & Zhao, F. (2005). Establishment of a micro-particle bombardment transformation system for Dunaliella salina. Journal of Microbiology, 43: 361–365.
  • Te, M.R., Lohuis & Miller, D.J. (1998). Genetic transformation of dinoflagellates (Amphidinium and Symbiodinium): expression of GUS in microalgae using heterologous promoter constructs. The Plant Journal, 13: 427–435.
  • Tenaillon, O., Rodriguez-Verdugo, A., Gaut, R.L., McDonald, P., Bennett, A.F., Long, A.D. & Gaut, B.S. (2012). The molecular diversity of adaptive convergence. Science, 335: 457–461.
  • Teng, C., Qin, S., Liu, J., Yu, D., Liang, C. & Tseng, C. (2002). Transient expression of lacZ in bombarded unicellular green alga Haematococcus pluvialis. Journal of Applied Phycology, 14: 495–500.
  • Tillich, U.M., Wolter. N., Franke, P., Dühring, U. & Frohme, M. (2014). Screening and genetic characterization of thermo-tolerant Synechocystis sp. PCC6803 strains created by adaptive evolution. BMC Biotechnology, 14: 66.
  • Tiwari, M., Sharma, D. & Trivedi, P.K. (2014). Artificial microRNA mediated gene silencing in plants: progress and perspectives. Plant Molecular Biology, 86: 1–18.
  • Tolonen, A.C., Liszt, G.B. & Hess, W.R. (2006). Genetic manipulation of Prochlorococcus strain MIT9313: green fluorescent protein expression from an RSF1010 plasmid and Tn5 transposition. Applied and Environmental Microbiology, 72: 7607–7613.
  • Toyomizu, M., Suzuki, K., Kawata, Y., Kojima, H. & Akiba, Y. (2001). Effective transformation of the cynobacterium Spirulina platensis using electroporation. Journal of Applied Phycology, 13: 209–214.
  • Tran, M., Zhou, B., Pettersson, P.L., Gonzalez, M.J. & Mayfield, S.P. (2009). Synthesis and assembly of a full-length human monoclonal antibody in algal chloroplasts. Biotechnology and Bioengineering, 104: 663–673.
  • 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: 569–576.
  • Uchiyama, J., Kanesaki, Y., Iwata, N., Asakura, R., Funamizu, K., Tasaki, R., Agatsuma, M., Tahara, H., Matsuhashi, A., Yoshikawa, H., Ogawa, S. & Ohta, H. (2015). Genomic analysis of parallel-evolved cyanobacterium Synechocystis sp. PCC 6803 under acid stress. Photosynthesis Research, 125: 243–254.
  • Uji, T., Takahashi, M., Saga, N. & Mikami, K. (2010). Visualization of nuclear localization of transcription factors with cyan and green fluorescent proteins in the red alga Porphyra yezoensis. Marine Biotechnology, 12: 150–159.
  • Uji, T., Hirata, R., Fukuda, S., Mizuta, H. & Saga, N. (2014). A codon-optimized bacterial antibiotic gene used as selection marker for stable nuclear transformation in the marine red alga Pyropia yezoensis. Marine Biotechnology, 16: 251–255.
  • Urnov, F.D., Miller, J.C., Lee, Y.L., Beausejour, C.M., Rock, J.M., Augustus, S., Jamieson, A.C., Porteus, M.H., Gregory, P.D. & Holmes, M.C. (2005). Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature, 435: 646–651.
  • 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: 636–646.
  • Velmurugan, N., Sung, M., Yim, S.S., Park, M.S., Yang, J.W. & Jeong, K.J. (2014). Systematically programmed adaptive evolution reveals potential role of carbon and nitrogen pathways during lipid accumulation in Chlamydomonas reinhardtii. Biotechnology for Biofuels, 7: 117.
  • Walsh, R.M. & Hochedlinger, K. (2013). A variant CRISPR-Cas9 system adds versatility to genome engineering. Proceedings of the National Academy of Sciences of the United States of America, 110: 15514–15515.
  • Wang, H., Yang, H., Shivalila, C.S., Dawlaty, M.M., Cheng, A.W., Zhang, F. & Jaenisch, R. (2013). One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell, 153: 910–918.
  • Wang, J., Jiang, P., Cui, Y., Deng, X., Li, F., Liu, J. & Qin, S. (2010a). Genetic transformation in Kappaphycus alvarezii using micro-particle bombardment: a potential strategy for germplasm improvement. Aquaculture International, 18: 1027–1034.
  • Wang, J., Jiang, P., Cui, Y., Guan, X. & Qin, S. (2010b). Gene transfer into conchospores of Porphyra haitanensis (Bangiales, Rhodophyta) by glass bead agitation. Phycologia, 49: 355–360.
  • Wang, Q., Lu, Y., Xin, Y., Wei, L., Huang, S. & Xu, J. (2016). Genome editing of model oleaginous microalgae Nannochloropsis spp. by CRISPR/Cas9. The Plant Journal, 88: 1071–1081.
  • Wang, Y., Shi, M., Niu, X., Zhang, X., Gao, L., Chen, L., Wang, J. & Zhang, W. (2014). Metabolomic basis of laboratory evolution of butanol tolerance in photosynthetic Synechocystis sp. PCC 6803. Microbial Cell Factories, 13: 151.
  • Wang, B., Wang, J., Zhang, W. & Meldrum D.R. (2012). Application of synthetic biology in cyanobacteria and algae. Frontiers in Microbiology, 3: 344.
  • Weber, E., Engler, C., Gruetzner, R., Werner, S. & Marillonnet, S. (2011). A modular cloning system for standardized assembly of multigene constructs. PLoS ONE, 6: e16765.
  • Wei, L., Xin, Y., Wang, Q., Yang, J., Hu, H. & Xu, J. (2017). RNAi-based targeted gene knockdown in the model oleaginous microalgae Nannochloropsis oceanica. The Plant Journal, 89: 1236–1250.
  • Weyman, P.D., Beeri, K., Lefebvre, S.C., Rivera, J., McCarthy, J.K., Heuberger, A.L., Peers, G., Allen, A.E. & Dupont, C.L. (2015). Inactivation of Phaeodactylum tricornutum urease gene using transcription activator-like effector nuclease-based targeted mutagenesis. Plant Biotechnology Journal, 13: 460–470.
  • Wu, C.Y. & Lin, G.H. (1987). Progress in the genetics and breeding of economic seaweeds in China. Hydrobiology, 151/152: 57–61.
  • Xie, S., Shen, B., Zhang, C., Huang, X. & Zhang, Y. (2014). sgRNAcas9: A software package for designing CRISPR sgRNA and evaluating potential off-target cleavage sites. PLoS ONE, 9: e100448.
  • Xing, H.L., Dong, L., Wang, Z.P., Zhang, H.Y., Han, C.Y., Liu, B., Wang, X.C. & Chen, Q.J. (2014). A CRISPR/Cas9 toolkit for multiplex genome editing in plants. BMC Plant Biology, 14: 327.
  • Xu, P., Zhang, Y., Kang, L., Roossinck, M.J. & Mysore, K.S. (2006). Computational estimation and experimental verification of off-target silencing during posttranscriptional gene silencing in plants. Plant Physiology, 142: 429–440.
  • Yamasaki, T., Miyasaka, H. & Ohama, T. (2008). Unstable RNAi effects through epigenetic silencing of an inverted repeat transgene in Chlamydomonas reinhardtii. Genetics, 180: 1927–1944.
  • Yi, Z., Xu, M., Magnusdottir, M., Zhang, Y., Brynjolfsson, S. & Fu, W. (2015). Photo-oxidative stress-driven mutagenesis and adaptive evolution on the marine diatom Phaeodactylum tricornutum for enhanced carotenoid accumulation. Marine Drugs, 13: 6138–6151.
  • Yu, C., Liu, Y., Ma, T., Liu, K., Xu, S., Zhang, Y., Liu, H., La Russa, M., Xie, M., Ding, S. & Qi, L.S. (2015). Small molecules enhance CRISPR genome editing in pluripotent stem cells. Cell Stem Cell, 16: 142–147.
  • Yu, D.Z., Qin S., Sun G.Q. & Tzeng C.K. (2002). Transient expression of lacZ reporter gene in the economic seaweed Undaria pinnatifida. High Technology Letters, 12: 93–95.
  • Yu, S., Zhao, Q., Miao, X. & Shi, J. (2013). Enhancement of lipid production in low-starch mutants Chlamydomonas reinhardtii by adaptive laboratory evolution. Bioresource Technology, 147: 499–507.
  • Zaslavskaia, L.A., Lippmeier, J.C., Kroth, P.G., Grossman, A.R. & Apt, K.E. (2000). Transformation of the diatom Phaeodactylum tricornutum (Bacillariophyceae) with a variety of selectable marker and reporter genes. Journal of Phycology, 36: 379–386.
  • Zhang, Y., Jiang, P., Gao, J., Liao, J., Sun, S., Shen, Z. & Qin, S. (2008). Recombinant expression of rt-PA gene (encoding Reteplase) in gametophytes of the seaweed Laminaria japonica (Laminariales, Phaeophyta). Science in China Series C-Life Sciences, 51: 1116–1120.
  • Zhang, F., Wen, Y. & Guo, X. (2014). CRISPR/Cas9 for genome editing: progress, implications and challenges. Human Molecular Genetics, 23: R40–R46.
  • Zhao, T., Li, G., Mi, S., Li, S., Hannon, G.J., Wang, X.J. & Qi, Y. (2007). A complex system of small RNAs in the unicellular green alga Chlamydomonas reinhardtii. Genes and Development, 21: 1190–1203.
  • Zhao, T., Wang, W., Bai, X. & Qi, Y. (2009). Gene silencing by artificial microRNAs in Chlamydomonas. The Plant Journal, 58: 157–164.
  • Zorin, B., Grundman, O., Khozin-Goldberg, I., Leu, S., Shapira, M., Kaye, Y., Tourasse, N., Vallon, O. & Boussiba, S. (2014). Development of a nuclear transformation system for oleaginous green alga Lobosphaera (Parietochloris) incisa and genetic complementation of a mutant strain, deficient in arachidonic acid biosynthesis. PLoS ONE, 9: e105223.
  • Zuo, Z., Li, B., Wang, C., Cai, J. & Chen, Y. (2007). Increasing transient expression of CAT gene in Porphyra haitanensis by matrix attachment regions and 18S rDNA targeted homologous recombination. Aquaculture Research, 38: 681–688.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.