S. Sasso, G. Pohnert, M. Lohr, M. Mittag, and C. Hertweck, Microalgae in the postgenomic era: a blooming reservoir for new natural products, FEMS Microbiology Reviews, vol.158, issue.4, pp.761-785, 2012.
DOI : 10.1146/annurev.arplant.59.032607.092730

P. Spolaore, C. Joannis-cassan, E. Duran, and A. Isambert, Commercial applications of microalgae, Journal of Bioscience and Bioengineering, vol.101, issue.2, pp.87-96, 2006.
DOI : 10.1263/jbb.101.87

URL : https://hal.archives-ouvertes.fr/hal-00133263

T. Mata, A. Martins, and N. Caetano, Microalgae for biodiesel production and other applications: A review. Renewable and Sustainable Energy Reviews, pp.217-232, 2010.
DOI : 10.1016/j.rser.2009.07.020

URL : http://recipp.ipp.pt/bitstream/10400.22/10059/1/ART_NidiaCaetano_DEQ_2009.pdf

J. Cadoret, M. Garnier, and B. Saint-jean, Microalgae, functional genomics and biotechnology . Advances in Botanical Research, pp.285-341, 2012.
DOI : 10.1016/b978-0-12-391499-6.00008-6

R. León-bañares, D. González-ballester, A. Galván, and E. Fernández, Transgenic microalgae as green cell-factories, Trends in Biotechnology, vol.22, issue.1, pp.45-52, 2004.
DOI : 10.1016/j.tibtech.2003.11.003

D. Barrera and S. Mayfield, High-value recombinant protein production in microalgae Handbook of Microalgal Culture [Internet]
DOI : 10.1002/9781118567166.ch27

F. Hempel and U. Maier, Microalgae as Solar-Powered Protein Factories, Advances in Experimental Medicine and Biology, vol.896, pp.241-262, 2016.
DOI : 10.1007/978-3-319-27216-0_16

S. Rosales-mendoza, Algae-Made Antibodies and Immunotoxins Available from: https, Algae-Based Biopharmaceuticals, vol.10, pp.77-93978, 1007.
DOI : 10.1007/978-3-319-32232-2_5

M. Tran, B. Zhou, P. Pettersson, M. Gonzalez, and S. Mayfield, Synthesis and assembly of a full-length human monoclonal antibody in algal chloroplasts, Biotechnology and Bioengineering, vol.16, issue.4, pp.663-673, 2009.
DOI : 10.1042/bj2590347

M. Tran, R. Henry, D. Siefker, C. Van, G. Newkirk et al., Production of anti-cancer immunotoxins in algae: Ribosome inactivating proteins as fusion partners, Biotechnology and Bioengineering, vol.278, issue.23, pp.2826-2835, 2013.
DOI : 10.1111/j.1742-4658.2011.08182.x

M. Tran, C. Van, D. Barrera, P. Pettersson, C. Peinado et al., Production of unique immunotoxin cancer therapeutics in algal chloroplasts, Proceedings of the National Academy of Sciences of the United States of America, pp.15-22, 2013.
DOI : 10.1371/journal.pone.0037179

URL : http://www.pnas.org/content/110/1/E15.full.pdf

F. Hempel, J. Lau, A. Klingl, and U. Maier, Algae as Protein Factories: Expression of a Human Antibody and the Respective Antigen in the Diatom Phaeodactylum tricornutum, PLoS ONE, vol.252, issue.12, p.28424, 2011.
DOI : 10.1371/journal.pone.0028424.g005

F. Hempel and U. Maier, An engineered diatom acting like a plasma cell secreting human IgG antibodies with high efficiency. Microbial Cell Factories, p.126, 2012.
DOI : 10.1186/1475-2859-11-126

URL : http://doi.org/10.1186/1475-2859-11-126

F. Hempel, M. Maurer, B. Brockmann, C. Mayer, N. Biedenkopf et al., From hybridomas to a robust microalgal-based production platform: molecular design of a diatom secreting monoclonal antibodies directed against the Marburg virus nucleoprotein, Microbial Cell Factories, vol.83, issue.Suppl 2, p.131, 2017.
DOI : 10.1016/j.jpba.2013.04.030

G. Vanier, F. Hempel, P. Chan, M. Rodamer, D. Vaudry et al., Biochemical Characterization of Human Anti-Hepatitis B Monoclonal Antibody Produced in the Microalgae Phaeodactylum tricornutum, PLOS ONE, vol.22, issue.10, p.139282, 2015.
DOI : 10.1371/journal.pone.0139282.t001

G. Vanier, S. Stelter, J. Vanier, F. Hempel, U. Maier et al., Alga-Made Anti-Hepatitis B Antibody Binds to Human Fc?? Receptors, Biotechnology Journal, vol.9, issue.4
DOI : 10.1002/pmic.200900708

URL : https://hal.archives-ouvertes.fr/hal-01777650/file/2017-Vanier_et_al-Biotech_J.pdf

O. Flaherty, R. Trbojevi?-akma?i?, I. Greville, G. Rudd, P. Lauc et al., The sweet spot for biologics: recent advances in characterization of biotherapeutic glycoproteins, Expert Review of Proteomics, vol.7, issue.13397, pp.13-29, 2018.
DOI : 10.1021/ac1013139

N. Lingg, P. Zhang, Z. Song, and M. Bardor, The sweet tooth of biopharmaceuticals: Importance of recombinant protein glycosylation analysis, Biotechnology Journal, vol.849, issue.1, pp.1462-1472, 2012.
DOI : 10.1016/j.jchromb.2006.09.041

URL : https://hal.archives-ouvertes.fr/hal-01844671

K. Brorson and A. Jia, Therapeutic monoclonal antibodies and consistent ends: terminal heterogeneity, detection, and impact on quality, Current Opinion in Biotechnology, vol.30, pp.140-146, 2014.
DOI : 10.1016/j.copbio.2014.06.012

B. Beyer, M. Schuster, A. Jungbauer, and N. Lingg, Microheterogeneity of Recombinant Antibodies: Analytics and Functional Impact, Biotechnology Journal, vol.11, issue.1, p.28862393, 2018.
DOI : 10.1002/biot.201600504

URL : http://onlinelibrary.wiley.com/doi/10.1002/biot.201700476/pdf

M. Van-beers and M. Bardor, Minimizing immunogenicity of biopharmaceuticals by controlling critical quality attributes of proteins, Biotechnology Journal, vol.10, issue.12, pp.1473-1484
DOI : 10.2174/138920109788488914

URL : https://hal.archives-ouvertes.fr/hal-01844668

P. Burda and M. Aebi, The dolichol pathway of N-linked glycosylation, BBA)?General Subjects, pp.239-257, 1999.
DOI : 10.1016/S0304-4165(98)00127-5

E. Weerapana and B. Imperiali, Asparagine-linked protein glycosylation: from eukaryotic to prokaryotic systems, Glycobiology, vol.16, issue.6, pp.91-101, 2006.
DOI : 10.1074/jbc.M206114200

G. Gil, W. Velander, and K. Van-cott, N-glycosylation microheterogeneity and site occupancy of an Asn-X-Cys sequon in plasma-derived and recombinant protein C, PROTEOMICS, vol.128, issue.9, pp.2555-2567, 2009.
DOI : 10.1093/oxfordjournals.jbchem.a022738

D. Zielinska, F. Gnad, J. Wi?niewski, and M. Mann, Precision mapping of an in vivo N-Glycoproteome reveals rigid topological and sequence constraints. Cell, pp.897-907, 2010.

T. Matsui, E. Takita, T. Sato, S. Kinjo, M. Aizawa et al., N-glycosylation at noncanonical Asn-X-Cys sequences in plant cells, Glycobiology, vol.28, issue.12, pp.994-999, 2011.
DOI : 10.3168/jds.2008-1189

D. Williams, Beyond lectins: the calnexin/calreticulin chaperone system of the endoplasmic reticulum, Journal of Cell Science, vol.119, issue.4, pp.615-623, 2006.
DOI : 10.1242/jcs.02856

A. Helenius and M. Aebi, Intracellular functions of N-linked glycans. Science, pp.2364-2369, 2001.

A. Varki, Loss of N-glycolylneuraminic acid in humans: Mechanisms, consequences, and implications for hominid evolution, American Journal of Physical Anthropology, vol.269, issue.S33, pp.54-69, 2001.
DOI : 10.1292/jvms1939.52.567

P. Lerouge, M. Cabanes-macheteau, C. Rayon, A. Fischette-lainé, V. Gomord et al., N-Glycoprotein biosynthesis in plants: recent developments and future trends, Plant Molecular Biology, vol.38, issue.12, pp.31-48, 1998.
DOI : 10.1007/978-94-011-5298-3_2

I. Wilson, R. Zeleny, D. Kolarich, E. Staudacher, C. Stroop et al., Analysis of Asn-linked glycans from vegetable foodstuffs: widespread occurrence of Lewis a, core ??1,3-linked fucose and xylose substitutions, Glycobiology, vol.51, issue.2, pp.261-274, 2001.
DOI : 10.1016/S0031-9422(98)00764-X

P. Gagneux and A. Varki, Evolutionary considerations in relating oligosaccharide diversity to biological function, Glycobiology, vol.99, issue.1, pp.747-755, 1999.
DOI : 10.1093/oxfordjournals.jbchem.a135479

A. Varki, R. Cummings, J. Esko, H. Freeze, P. Stanley et al., Available from, Essentials of Glycobiology [Internet]. 2nd ed. Cold Spring Harbor (NY), 1908.

D. Balshüsemann and L. Jaenicke, The oligosaccharides of the glycoprotein pheromone of Volvox carteri f. nagariensis Iyengar (Chlorophyceae), European Journal of Biochemistry, vol.5, issue.1, pp.231-237, 1990.
DOI : 10.1016/0014-5793(81)80938-6

A. Grunow, B. Becker, and M. Melkonian, Isolation and characterization of the Golgi apparatus of a flagellate scaly green alga, European Journal of Cell Biology, vol.61, issue.1, pp.10-20, 1993.

S. Gödel, B. Becker, and M. Melkonian, Flagellar Membrane Proteins of Tetraselmis striata Butcher (Chlorophyta), Protist, vol.151, issue.2, pp.147-159, 2000.
DOI : 10.1078/1434-4610-00015

O. Levy-ontman, S. Arad, D. Harvey, T. Parsons, A. Fairbanks et al., sp., Journal of Biological Chemistry, vol.271, issue.Part 3, pp.21340-21352, 2011.
DOI : 10.1093/glycob/7.5.663

O. Levy-ontman, Glycosylation of the 66-kDa Cell-Wall Glycoprotein of a Red Microalga Available from: http://www.intechopen.com/books/ glycosylation/n-glycosylation-of-the-66-kda-cell-wall-glycoprotein-of-a-red-microalga, 2012.

E. Mathieu-rivet, M. Scholz, C. Arias, F. Dardelle, S. Schulze et al., Unravels Novel Complex Structures, Molecular & Cellular Proteomics, vol.2, issue.11, pp.3160-3183, 2013.
DOI : 10.1002/pmic.200900708

URL : https://hal.archives-ouvertes.fr/hal-00996460

G. Vanier, P. Lucas, C. Loutelier-bourhis, J. Vanier, C. Plasson et al., Heterologous expression of the N-acetylglucosaminyltransferase I dictates a reinvestigation of the N-glycosylation pathway in Chlamydomonas reinhardtii Scientific Reports, p.10156, 2017.

B. Baïet, C. Burel, B. Saint-jean, R. Louvet, L. Menu-bouaouiche et al., N-Glycans of Phaeodactylum tricornutum diatom and functional characterization of its N-acetylglucosaminyltransferase I enzyme, The Journal of Biological Chemistry. Feb, vol.25286, issue.8, pp.6152-6164, 2011.

S. Schulze, E. Urzica, M. Reijnders, H. Van-de-geest, S. Warris et al., Identification of methylated GnTI-dependent N-glycans in Botryococcus brauni. The New Phytologist, pp.1361-1369, 2017.
DOI : 10.1111/nph.14713

S. Casabianca, L. Cornetti, S. Capellacci, C. Vernesi, and A. Penna, Genome complexity of harmful microalgae. Harmful Algae, pp.7-12, 2017.
DOI : 10.1016/j.hal.2017.01.003

O. Levy-ontman, M. Fisher, Y. Shotland, Y. Weinstein, Y. Tekoah et al., Genes Involved in the Endoplasmic Reticulum N-Glycosylation Pathway of the Red Microalga Porphyridium sp.: A Bioinformatic Study, International Journal of Molecular Sciences, vol.24, issue.2, pp.2305-2326, 2014.
DOI : 10.1093/molbev/msm092

E. Mathieu-rivet, M. Kiefer-meyer, G. Vanier, C. Ovide, C. Burel et al., Protein N-glycosylation in eukaryotic microalgae and its impact on the production of nuclear expressed biopharmaceuticals, Available from, 2014.
DOI : 10.1074/jbc.270.29.17344

S. Kallolimath, A. Castilho, R. Strasser, C. Grünwald-gruber, F. Altmann et al., Engineering of complex protein sialylation in plants, Proceedings of the National Academy of Sciences of the United States of America, pp.9498-9503, 2016.
DOI : 10.1006/niox.2000.0319

URL : http://www.pnas.org/content/113/34/9498.full.pdf

S. Hamilton, R. Davidson, N. Sethuraman, J. Nett, Y. Jiang et al., Humanization of Yeast to Produce Complex Terminally Sialylated Glycoproteins, Science, vol.24, issue.2, pp.3131441-1443, 2006.
DOI : 10.1038/nbt1178

B. Gügi, L. Costaouec, T. Burel, C. Lerouge, P. Helbert et al., Diatom-specific oligosaccharide and polysaccharide structures help to unravel biosynthetic capabilities in diatoms. Marine Drugs, pp.5993-6018, 2015.

E. Mathieu-rivet, P. Lerouge, and M. Bardor, Chlamydomonas reinhardtii: Protein Glycosylation and Production of Biopharmaceuticals, Chlamydomonas: Biotechnology and Biomedicine. Microbiology Monographs. Cham, vol.1, issue.2, pp.978-981
DOI : 10.4155/pbp.13.7

A. Galván, D. González-ballester, and E. Fernández, Insertional Mutagenesis as a Tool to Study Genes/Functions in Chlamydomonas, Advances in Experimental Medicine and Biology, vol.616, pp.77-89, 2007.
DOI : 10.1007/978-0-387-75532-8_7

L. Doron, N. Segal, and M. Shapira, Transgene Expression in Microalgae???From Tools to Applications, Frontiers in Plant Science, vol.4, issue.597, p.505, 2016.
DOI : 10.1128/EC.4.7.1264-1272.2005

URL : http://journal.frontiersin.org/article/10.3389/fpls.2016.00505/pdf

F. Daboussi, Advances in editing microalgae genomes, Perspectives in Phycology, vol.4, issue.1, pp.17-23, 2017.
DOI : 10.1127/pip/2017/0071

URL : https://hal.archives-ouvertes.fr/hal-01606205

I. Ng, S. Tan, P. Kao, Y. Chang, and J. Chang, Recent Developments on Genetic Engineering of Microalgae for Biofuels and Bio-Based Chemicals, Biotechnology Journal, vol.38, issue.10, p.1600644, 2017.
DOI : 10.1007/PL00006813

A. Falciatore, R. Casotti, C. Leblanc, C. Abrescia, and C. Bowler, Transformation of Nonselectable Reporter Genes in Marine Diatoms, Marine Biotechnology, vol.1, issue.3, pp.239-251, 1999.
DOI : 10.1007/PL00011773

K. Kindle, High-frequency nuclear transformation of Chlamydomonas reinhardtii, Proceedings of the National Academy of Sciences, pp.1228-1232, 1990.
DOI : 10.1073/pnas.87.3.1228

URL : http://www.pnas.org/content/87/3/1228.full.pdf

E. Armbrust, Identification of a new gene family expressed during the onset of sexual reproduction in the centric DiatomThalassiosira weissflogii, Applied and Environmental Microbiology, vol.65, issue.7, pp.3121-3128, 1999.

M. Takata, M. Sasaki, E. Sonoda, C. Morrison, M. Hashimoto et al., Homologous recombination and non-homologous end-joining pathways of DNA double-strand break repair have overlapping roles in the maintenance of chromosomal integrity in vertebrate cells, The EMBO Journal, vol.17, issue.18, pp.5497-5508, 1998.
DOI : 10.1093/emboj/17.18.5497

M. Maeder and C. Gersbach, Genome-editing technologies for gene and cell therapy. Molecular Therapy, pp.430-446, 2016.
DOI : 10.1038/mt.2016.10

URL : https://doi.org/10.1038/mt.2016.10

F. Daboussi, S. Leduc, A. Maréchal, G. Dubois, V. Guyot et al., Genome engineering empowers the diatom Phaeodactylum tricornutum for biotechnology, Nature Communications, vol.942, issue.943, p.3831, 2014.
DOI : 10.1016/j.jchromb.2013.10.016

URL : https://hal.archives-ouvertes.fr/hal-01528413

J. Miller, M. Holmes, J. Wang, D. Guschin, Y. Lee et al., An improved zinc-finger nuclease architecture for highly specific genome editing, Nature Biotechnology, vol.11, issue.7, p.1319, 2007.
DOI : 10.1038/nbt1319

J. Smith, M. Bibikova, F. Whitby, A. Reddy, S. Chandrasegaran et al., Requirements for double-strand cleavage by chimeric restriction enzymes with zinc finger DNA-recognition domains, Nucleic Acids Research, vol.28, issue.17, pp.3361-3369, 2000.
DOI : 10.1093/nar/28.17.3361

URL : http://europepmc.org/articles/pmc110700?pdf=render

I. Sizova, A. Greiner, M. Awasthi, S. Kateriya, and P. Hegemann, Nuclear gene targeting in Chlamydomonas using engineered zinc-finger nucleases. The Plant Journal, pp.873-882, 2013.
DOI : 10.1111/tpj.12066

URL : http://onlinelibrary.wiley.com/doi/10.1111/tpj.12066/pdf

A. Greiner, S. Kelterborn, H. Evers, G. Kreimer, I. Sizova et al., Targeting of Photoreceptor Genes in Chlamydomonas reinhardtii via Zinc-finger Nucleases and CRISPR/Cas9, Plant Cell, 2017.
DOI : 10.1105/tpc.17.00659

R. Gabriel, A. Lombardo, A. Arens, J. Miller, P. Genovese et al., An unbiased genome-wide analysis of zinc-finger nuclease specificity, Nature Biotechnology, vol.649, issue.9, 1948.
DOI : 10.1006/jmbi.1990.9999

M. Christian, T. Cermak, E. Doyle, C. Schmidt, F. Zhang et al., Targeting DNA Double-Strand Breaks with TAL Effector Nucleases, Genetics, vol.186, issue.2, pp.757-761, 2010.
DOI : 10.1534/genetics.110.120717

URL : http://www.genetics.org/content/genetics/186/2/757.full.pdf

S. Kay, S. Hahn, E. Marois, G. Hause, and U. Bonas, A bacterial effector acts as a plant transcription factor and induces a cell size regulator. Science, pp.318648-651, 2007.
DOI : 10.1126/science.1144956

P. Weyman, K. Beeri, S. Lefebvre, J. Rivera, J. Mccarthy et al., urease gene using transcription activator-like effector nuclease-based targeted mutagenesis, Plant Biotechnology Journal, vol.333, issue.125, pp.460-470, 2015.
DOI : 10.1126/science.1207773

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1111/pbi.12254

A. Fortunato, M. Jaubert, G. Enomoto, J. Bouly, R. Raniello et al., Diatom Phytochromes Reveal the Existence of Far-Red-Light-Based Sensing in the Ocean, The Plant Cell, vol.28, issue.3, 2016.
DOI : 10.1105/tpc.15.00928

URL : https://hal.archives-ouvertes.fr/hal-01528502

H. Liao, F. Hatanaka, T. Araoka, P. Reddy, M. Wu et al., In??Vivo Target Gene Activation via CRISPR/Cas9-Mediated Trans -epigenetic Modulation, Cell, vol.171, issue.7, pp.92-867431247, 2009.
DOI : 10.1016/j.cell.2017.10.025

K. Makarova, D. Haft, R. Barrangou, S. Brouns, E. Charpentier et al., Evolution and classification of the CRISPR???Cas systems, Nature Reviews Microbiology, vol.35, issue.6, pp.467-477, 2011.
DOI : 10.1093/nar/gkl1043

URL : http://europepmc.org/articles/pmc3380444?pdf=render

M. Jinek, K. Chylinski, I. Fonfara, M. Hauer, J. Doudna et al., A programmable dual-RNA?guided DNA endonuclease in adaptive bacterial immunity. Science, pp.816-821, 2012.
DOI : 10.1126/science.1225829

URL : https://zenodo.org/record/1230922/files/article.pdf

W. Jiang, A. Brueggeman, K. Horken, T. Plucinak, and D. Weeks, ABSTRACT, Eukaryotic Cell, vol.13, issue.11, pp.1465-1469, 2014.
DOI : 10.1128/EC.00213-14

S. Shin, J. Lim, H. Koh, E. Kim, N. Kang et al., CRISPR/Cas9-induced knockout and knock-in mutations in Chlamydomonas reinhardtii Scientific Reports, p.27810, 2016.
DOI : 10.1038/srep27810

URL : http://www.nature.com/articles/srep27810.pdf

M. Nymark, A. Sharma, T. Sparstad, A. Bones, and P. Winge, A CRISPR/Cas9 system adapted for gene editing in marine algae Scientific Reports, p.24951, 2016.

A. Hopes, V. Nekrasov, S. Kamoun, and T. Mock, Editing of the urease gene by CRISPR-Cas in the diatom Thalassiosira pseudonana. Plant Methods, p.49, 2016.

Q. Wang, Y. Lu, Y. Xin, L. Wei, S. Huang et al., Genome editing of model oleaginous microalgae Nannochloropsis spp. by CRISPR/Cas9. The Plant Journal, pp.1071-1081, 2016.

B. Chevalier and B. Stoddard, Homing endonucleases: structural and functional insight into the catalysts of intron/intein mobility, Nucleic Acids Research, vol.29, issue.18, pp.3757-3774, 2001.
DOI : 10.1093/nar/29.18.3757

URL : https://academic.oup.com/nar/article-pdf/29/18/3757/9906071/293757.pdf

J. Joung and J. Sander, TALENs: a widely applicable technology for targeted genome editing, Nature Reviews Molecular Cell Biology, vol.15, issue.1, pp.49-55, 2013.
DOI : 10.1093/nar/gks608

URL : http://europepmc.org/articles/pmc3547402?pdf=render

X. Li, R. Zhang, W. Patena, S. Gang, S. Blum et al., An indexed, mapped mutant library enables reverse genetics studies of biological processes in Chlamydomonas reinhardtii . The Plant Cell, Feb, vol.28, issue.2, pp.367-387, 2016.
DOI : 10.1105/tpc.15.00465

URL : http://www.plantcell.org/content/plantcell/28/2/367.full.pdf

, Toward Future Engineering of the N-Glycosylation Pathways in Microalgae for Optimizing the Production? http