F. Altmann, H. Schwihla, E. Staudacher, J. Glössl, and L. März, Insect cells contain an unusual, membrane-bound-N-acetylglucosaminidase probably involved in the processing of protein N-glycans, J. Biol. Chem, vol.270, pp.17344-17349, 1995.

E. V. Ambrust, J. Berges, C. Bowler, B. Green, D. Martinez et al., The Genome of the Diatom Thalassiosira Pseudonana: Ecology, Evolution, and Metabolism, 2004.

K. E. Apt, A. R. Grossman, and P. G. Kroth-pancic, Stable nuclear transformation of the diatomPhaeodactylum tricornutum, Mol. Gen. Genet, vol.252, pp.572-579, 1996.

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, J. Biol. Chem, vol.286, pp.6152-6164, 2011.

L. Bai, W. Yin, Y. Chen, L. Niu, Y. Sun et al., A new strategy to produce a defensin: stable production of mutated NP-1 in nitrate reductase-deficient chlorella ellipsoidea, PLoS ONE, vol.8, p.54966, 2013.

H. Bakker, M. Bardor, J. W. Molthoff, V. Gomord, I. Elbers et al., Galactose-extended glycans of antibodies produced by transgenic plants, Proc. Natl. Acad. Sci. U.S.A, vol.98, pp.2899-2904, 2001.

H. Bakker, G. J. Rouwendal, A. S. Karnoup, D. E. Florack, G. M. Stoopen et al., An antibody produced in tobacco expressing a hybrid ?-1, 4-galactosyltransferase is essentially devoid of plant carbohydrate epitopes, Proc. Natl. Acad. Sci. U.S.A, vol.103, pp.7577-7582, 2006.

D. Balshüsemann, J. , and L. , The oligosaccharides of the glycoprotein pheromone of Volvox carteri f. nagariensis Iyengar (Chlorophyceae), Eur. J. Biochem, vol.192, pp.231-237, 1990.

M. Bardor, C. Faveeuw, A. Fitchette, D. Gilbert, L. Galas et al., Immunoreactivity in mammals of two typical plant glyco-epitopes, core ? (1,3)-fucose and core xylose, Glycobiology, vol.13, pp.427-434, 2003.

L. L. Beer, E. S. Boyd, J. W. Peters, and M. C. Posewitz, Engineering algae for biohydrogen and biofuel production, Curr. Opin. Biotechnol, vol.20, pp.264-271, 2009.

G. Blanc, I. Agarkova, J. Grimwood, A. Kuo, A. Brueggeman et al., The genome of the polar eukaryotic microalga Coccomyxa subellipsoidea reveals traits of cold adaptation, Genome Biol, vol.13, p.39, 2012.

G. Blanc, G. Duncan, I. Agarkova, M. Borodovsky, J. Gurnon et al., The Chlorella variabilis NC64A genome reveals adaptation to photosymbiosis, coevolution with viruses, and cryptic sex, Plant Cell Online, vol.22, pp.2943-2955, 2010.

C. Bogen, A. Al-dilaimi, A. Albersmeier, J. Wichmann, M. Grundmann et al., Reconstruction of the lipid metabolism for the microalga Monoraphidium neglectum from its genome sequence reveals characteristics suitable for biofuel production, BMC Genomics, vol.14, p.926, 2013.

P. Burda, A. , and M. , The dolichol pathway of N-linked glycosylation, Biochim. Biophys. Acta, vol.1426, pp.127-132, 1999.

J. F. Buyel and R. Fischer, Downstream processing of biopharmaceutical proteins produced in plants: the pros and cons of flocculants, Bioengineered, vol.5, pp.138-142, 2014.

J. Cadoret and O. Bernard, La production de biocarburant lipidique avec des microalgues: promesses et défis, J. Soc. Biol, vol.202, pp.201-211, 2008.

A. Castilho, L. Neumann, P. Gattinger, R. Strasser, K. Vorauer-uhl et al., Generation of biologically active multi-sialylated recombinant human EPOFc in plants, PLoS ONE, vol.8, p.54836, 2013.

A. Castilho, M. Pabst, R. Leonard, C. Veit, F. Altmann et al., Construction of a functional CMP-sialic acid biosynthesis pathway in Arabidopsis, Plant Physiol, vol.147, pp.331-339, 2008.

A. Castilho, R. Strasser, J. Stadlmann, J. Grass, J. Jez et al., In planta protein sialylation through overexpression of the respective mammalian pathway, J. Biol. Chem, vol.285, pp.15923-15930, 2010.

H. Cerutti, A. M. Johnson, N. W. Gillham, and J. E. Boynton, Epigenetic silencing of a foreign gene in nuclear transformants of Chlamydomonas, Plant Cell, vol.9, pp.925-945, 1997.

H. Cerutti, X. Ma, J. Msanne, and T. Repas, RNA-mediated silencing in algae: biological roles and tools for analysis of gene function, Eukaryot Cell, vol.10, pp.1164-1172, 2011.

X. Chai, H. Chen, W. Xu, and Y. Xu, Expression of soybean Kunitz trypsin inhibitor gene SKTI in Dunaliella salina, J. Appl. Phycol, vol.25, pp.139-144, 2013.

Y. Chen, Y. Wang, Y. Sun, L. Zhang, L. et al., Highly efficient expression of rabbit neutrophil peptide-1 gene in Chlorella ellipsoidea cells, Curr. Genet, vol.39, pp.365-370, 2001.

C. H. Chung, B. Mirakhur, E. Chan, Q. Le, J. Berlin et al., , 2008.

, Cetuximab-induced anaphylaxis and IgE specific for galactose-?-1, 3-galactose. New Engl, J. Med, vol.358, pp.1109-1117

R. Colgan, C. J. Atkinson, M. Paul, S. Hassan, P. M. Drake et al., Optimisation of contained Nicotiana tabacum cultivation for the production of recombinant protein pharmaceuticals, Transgenic Res, vol.19, pp.241-256, 2010.

K. M. Cox, J. D. Sterling, J. T. Regan, J. R. Gasdaska, K. K. Frantz et al., Glycan optimization of a human monoclonal antibody in the aquatic plant Lemna minor, Nat. Biotechnol, vol.24, pp.1591-1597, 2006.

M. Crispin, D. J. Harvey, V. T. Chang, C. Yu, A. R. Aricescu et al., Inhibition of hybrid-and complex-type glycosylation reveals the presence of the GlcNAc transferase I-independent fucosylation pathway, Glycobiology, vol.16, pp.748-756, 2006.

E. L. Decker and R. Reski, Glycoprotein production in moss bioreactors, Plant Cell Rep, vol.31, pp.453-460, 2012.

A. L. Demain and P. Vaishnav, Production of recombinant proteins by microbes and higher organisms, Biotechnol. Adv, vol.27, pp.297-306, 2009.

O. C. Demurtas, S. Massa, P. Ferrante, A. Venuti, R. Franconi et al., A chlamydomonas-derived human papillomavirus 16 E7 vaccine induces specific tumor protection, PLoS ONE, vol.8, 2013.

, Frontiers in Plant Science | Plant Physiology, vol.5, 2014.

, Plant Cells, pp.31-48, 2013.

O. Levy-ontman, S. M. Arad, D. J. Harvey, T. B. Parsons, A. Fairbanks et al., Unique N-glycan moieties of the 66-kDa cell wall glycoprotein from the red microalga Porphyridium sp, J. Biol. Chem, vol.286, pp.21340-21352, 2011.

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, Int. J. Mol. Sci, vol.15, pp.2305-2326, 2014.

N. Lingg, P. Zhang, Z. Song, and M. Bardor, The sweet tooth of biopharmaceuticals: importance of recombinant protein glycosylation analysis, Biotechnol. J, vol.7, pp.1462-1472, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01844671

V. Lumbreras, D. R. Stevens, and S. Purton, Efficient foreign gene expression in Chlamydomonas reinhardtii mediated by an endogenous intron, Plant J, vol.14, pp.441-447, 1998.

T. M. Mata, A. A. Martins, and N. S. Caetano, Microalgae for biodiesel production and other applications: a review, Renew. Sust. Energ. Rev, vol.14, pp.217-232, 2010.

E. Mathieu-rivet, M. Scholz, C. Arias, F. Dardelle, S. Schulze et al., Exploring the N-glycosylation pathway in chlamydomonas reinhardtii unravels novel complex structures, Mol. Cell. Proteomics, vol.12, pp.3160-3183, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00996460

T. Matsui, E. Takita, T. Sato, S. Kinjo, M. Aizawa et al., Nglycosylation at noncanonical Asn-X-Cys sequences in plant cells, Glycobiology, vol.21, pp.994-999, 2011.

A. Maxmen, Drug-making plant blooms, Nature, vol.485, pp.160-160, 2012.

S. P. Mayfield, S. E. Franklin, and R. A. Lerner, Expression and assembly of a fully active antibody in algae, Proc. Natl. Acad. Sci, vol.100, pp.438-442, 2003.

S. S. Merchant, J. Kropat, B. Liu, J. Shaw, and J. Warakanont, , 2012.

, re it! Chlamydomonas as a reference organism for understanding algal triacylglycerol accumulation, Curr. Opin. Biotechnol, vol.23, pp.352-363

S. S. Merchant, S. E. Prochnik, O. Vallon, E. H. Harris, S. J. Karpowicz et al., The Chlamydomonas genome reveals the evolution of key animal and plant functions, Science, vol.318, pp.245-250, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00188075

M. Metzler, A. Gertz, M. Sarkar, H. Schachter, J. W. Schrader et al., Complex asparagine-linked oligosaccharides are required for morphogenic events during post-implantation development, EMBO J, vol.13, pp.2056-2065, 1994.

R. Misaki, K. Fujiyama, and T. Seki, Expression of human CMP-Nacetylneuraminic acid synthetase and CMP-sialic acid transporter in tobacco suspension-cultured cell, Biochem. Biophys. Res. Commun, vol.339, pp.1184-1189, 2006.

O. Misumi, M. Matsuzaki, H. Nozaki, S. Miyagishima, T. Mori et al., Cyanidioschyzon merolae genome. A tool for facilitating comparable studies on organelle biogenesis in photosynthetic eukaryotes, Plant Physiol, vol.137, pp.567-585, 2005.

J. Neupert, D. Karcher, and R. Bock, Generation of Chlamydomonas strains that efficiently express nuclear transgenes, Plant J, vol.57, 2009.

T. Paccalet, M. Bardor, C. Rihouey, F. Delmas, C. Chevalier et al., Diversity in specificity, abundance, and composition of anti-Neu5Gc antibodies in normal humans: potential implications for disease, Plant Biotechnol. J, vol.5, pp.818-830, 2007.

N. Q. Palacpac, S. Yoshida, H. Sakai, Y. Kimura, K. Fujiyama et al., Stable expression of human ?1, 4-galactosyltransferase in plant cells modifies N-linked glycosylation patterns, Proc. Natl. Acad. Sci. U.S.A, vol.96, pp.4692-4697, 1999.

B. Palenik, J. Grimwood, A. Aerts, P. Rouzé, A. Salamov et al., The tiny eukaryote Ostreococcus provides genomic insights into the paradox of plankton speciation, Proc. Natl. Acad. Sci. U.S.A, vol.104, pp.7705-7710, 2007.

S. Park, Y. Lee, J. Lee, J. , and E. , Expression of the high lightinducible Dunaliella LIP promoter in Chlamydomonas reinhardtii, Planta, vol.238, pp.1147-1156, 2013.

J. Parsons, F. Altmann, C. K. Arrenberg, A. Koprivova, A. K. Beike et al., Moss-based production of asialo-erythropoietin devoid of Lewis A and other plant-typical carbohydrate determinants, Plant Biotechnol. J, vol.10, pp.851-861, 2012.

G. Potvin and Z. Zhang, Strategies for high-level recombinant protein expression in transgenic microalgae: a review, Biotechnol. Adv, vol.28, pp.910-918, 2010.

N. Poulsen and N. Kröger, A new molecular tool for transgenic diatoms, Febs J, vol.272, pp.3413-3423, 2005.

R. Radakovits, R. E. Jinkerson, S. I. Fuerstenberg, H. Tae, R. E. Settlage et al., Draft genome sequence and genetic transformation of the oleaginous alga Nannochloropis gaditana, Nat. Commun, vol.3, p.686, 2012.

B. A. Rasala, P. A. Lee, Z. Shen, S. P. Briggs, M. Mendez et al., Robust expression and secretion of Xylanase1 in chlamydomonas reinhardtii by fusion to a selection gene and processing with the FMDV 2A peptide, PLoS ONE, vol.7, p.43349, 2012.

B. A. Rasala and S. P. Mayfield, The microalga Chlamydomonas reinhardtii as a platform for the production of human protein therapeutics, vol.2, pp.50-54, 2011.

B. A. Rasala and S. P. Mayfield, Photosynthetic biomanufacturing in green algae; production of recombinant proteins for industrial, nutritional, and medical uses, Photosyn. Res, 2014.

B. A. Rasala, M. Muto, P. A. Lee, M. Jager, R. M. Cardoso et al., Production of therapeutic proteins in algae, analysis of expression of seven human proteins in the chloroplast of Chlamydomonas reinhardtii: production of therapeutic proteins in algae, Plant Biotechnol. J, vol.8, pp.719-733, 2010.

G. J. Rouwendal, M. Wuhrer, D. E. Florack, C. A. Koeleman, A. M. Deelder et al., Efficient introduction of a bisecting GlcNAc residue in tobacco N-glycans by expression of the gene encoding human Nacetylglucosaminyltransferase III, Glycobiology, vol.17, pp.334-344, 2007.

O. Ruecker, K. Zillner, R. Groebner-ferreira, and M. Heitzer, Gaussialuciferase as a sensitive reporter gene for monitoring promoter activity in the nucleus of the green alga Chlamydomonas reinhardtii, Mol. Genet. Genomics, vol.280, pp.153-162, 2008.

S. Sasso, G. Pohnert, M. Lohr, M. Mittag, and C. Hertweck, Microalgae in the postgenomic era: a blooming reservoir for new natural products, FEMS Microbiol. Rev, vol.36, pp.761-785, 2012.

M. Schähs, R. Strasser, J. Stadlmann, R. Kunert, T. Rademacher et al., Production of a monoclonal antibody in plants with a humanized N-glycosylation pattern, Plant Biotechnol. J, vol.5, pp.657-663, 2007.

S. Schillberg, N. Raven, R. Fischer, R. Twyman, and A. Schiermeyer, Molecular farming of pharmaceutical proteins using plant suspension cell and tissue cultures, Curr. Pharm. Des, vol.19, pp.5531-5542, 2013.

M. Schroda, D. Blöcker, and C. F. Beck, The HSP70A promoter as a tool for the improved expression of transgenes in Chlamydomonas, Plant J, vol.21, pp.121-131, 2000.

M. Séveno, M. Bardor, T. Paccalet, V. Gomord, P. Lerouge et al., Glycoprotein sialylation in plants?, Nat. Biotechnol, vol.22, pp.1351-1352, 2004.

Y. Shaaltiel, D. Bartfeld, S. Hashmueli, G. Baum, E. Brill-almon et al., Production of glucocerebrosidase with terminal mannose glycans for enzyme replacement therapy of Gaucher's disease using a plant cell system, Plant Biotechnol. J, vol.5, pp.579-590, 2007.

S. Shaver, J. A. Casas-mollano, R. L. Cerny, and H. Cerutti, Origin of the polycomb repressive complex 2 and gene silencing by an E(z) homolog in the unicellular alga Chlamydomonas, Epigenetics, vol.5, pp.301-312, 2010.

E. Specht, S. Miyake-stoner, and S. Mayfield, Micro-algae come of age as a platform for recombinant protein production, Biotechnol. Lett, vol.32, pp.1373-1383, 2010.

, Frontiers in Plant Science | Plant Physiology, vol.5, 2014.

P. Spolaore, C. Joannis-cassan, E. Duran, and A. Isambert, Commercial applications of microalgae, J. Biosci. Bioeng, vol.101, pp.87-96, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00133263

E. Staudacher, Methylation-an uncommon modification of glycans, Biol. Chem, vol.393, pp.675-685, 2012.

E. Stoger, J. K. Ma, .. Fischer, R. Christou, and P. , Sowing the seeds of success: pharmaceutical proteins from plants, Curr. Opin. Biotechnol, vol.16, pp.167-173, 2005.

S. Stork, D. Moog, J. M. Przyborski, I. Wilhelmi, S. Zauner et al., Distribution of the SELMA translocon in secondary plastids of red algal origin and predicted uncoupling of ubiquitin-dependent translocation from degradation, Eukaryotic Cell, vol.11, pp.1472-1481, 2012.

R. Strasser, J. Mucha, L. Mach, F. Altmann, I. B. Wilson et al., Molecular cloning and functional expression of beta1, 2-xylosyltransferase cDNA from Arabidopsis thaliana, FEBS Lett, vol.472, pp.1443-1448, 2000.

R. Strasser, J. Stadlmann, M. Schähs, G. Stiegler, H. Quendler et al., Generation of glyco-engineered Nicotiana benthamiana for the production of monoclonal antibodies with a homogeneous humanlike N-glycan structure, Plant Biotechnol. J, vol.6, pp.133-138, 2008.

M. Tran, C. Van, D. J. Barrera, P. L. Pettersson, C. D. Peinado et al., Production of unique immunotoxin cancer therapeutics in algal chloroplasts, Proc. Natl. Acad. Sci. U.S.A, vol.110, 2012.

M. Tran, B. Zhou, P. L. Pettersson, M. J. Gonzalez, and S. P. Mayfield, Synthesis and assembly of a full-length human monoclonal antibody in algal chloroplasts, Biotechnol. Bioeng, vol.104, pp.663-673, 2009.

R. Twyman, S. Schillberg, and R. Fischer, Optimizing the yield of recombinant pharmaceutical proteins in plants, Curr. Pharm. Des, vol.19, pp.5486-5494, 2013.

M. M. Van-beers and M. Bardor, Minimizing immunogenicity of biopharmaceuticals by controlling critical quality attributes of proteins, Biotechnol. J, vol.7, pp.1473-1484, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01844668

S. M. Van-patten, H. Hughes, M. R. Huff, P. A. Piepenhagen, J. Waire et al., Effect of mannose chain length on targeting of glucocerebrosidase for enzyme replacement therapy of Gaucher disease, Glycobiology, vol.17, pp.467-478, 2007.

A. Varki, Biological roles of oligosaccharides: all of the theories are correct, Glycobiology, vol.3, 1993.

A. Varki, Evolutionary forces shaping the Golgi glycosylation machinery: why cell surface glycans are universal to living cells, Cold Spring Harb. Perspect. Biol, vol.3, p.5462, 2011.

A. Varki, R. D. Cummings, J. D. Esko, H. H. Freeze, P. Stanley et al., Symbol nomenclature for glycan representation, Proteomics, vol.9, pp.5398-5399, 2009.

L. Vézina, L. Faye, P. Lerouge, M. D'aoust, E. Marquet-blouin et al., Transient co-expression for fast and high-yield production of antibodies with human-like N-glycans in plants, Plant Biotechnol. J, vol.7, pp.442-455, 2009.

A. Vieler, G. Wu, C. Tsai, B. Bullard, A. J. Cornish et al., , 2012.

, functional gene annotation, and nuclear transformation of the heterokont oleaginous alga nannochloropsis oceanica CCMP1779, PLoS Genet, vol.8, p.1003064

A. Vitale and M. J. Chrispeels, Transient N-acetylglucosamine in the biosynthesis of phytohemagglutinin: attachment in the Golgi apparatus and removal in protein bodies, J. Cell Biol, vol.99, pp.133-140, 1984.

G. Walsh, Biopharmaceutical benchmarks 2010, Nat. Biotechnol, vol.28, pp.917-924, 2010.

E. Weerapana and B. Imperiali, Asparagine-linked protein glycosylation: from eukaryotic to prokaryotic systems, Glycobiology, vol.16, pp.91-101, 2006.

N. J. Weinreb, Imiglucerase and its Use for the Treatment of Gaucher's Disease, 1987.

I. B. Wilson, R. Zeleny, D. Kolarich, E. Staudacher, C. J. 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.11, pp.261-274, 2001.

C. Wong, Protein glycosylation: new challenges and opportunities, J. Org. Chem, vol.70, pp.4219-4225, 2005.

A. Z. Worden, J. Lee, T. Mock, P. Rouze, M. P. Simmons et al., Green evolution and dynamic adaptations revealed by genomes of the marine picoeukaryotes micromonas, Science, vol.324, pp.268-272, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00693449

J. Xu, M. C. Dolan, G. Medrano, C. L. Cramer, and P. J. Weathers, Green factory: plants as bioproduction platforms for recombinant proteins, Biotechnol. Adv, vol.30, pp.1171-1184, 2012.

L. A. Zaslavskaia, J. C. Lippmeier, P. G. Kroth, A. R. Grossman, and K. E. Apt, Transformation of the diatom Phaeodactylum tricornutum (Bacillariophyceae) with a variety of selectable marker and reporter genes, J. Phycol, vol.36, pp.379-386, 2000.

R. Zeleny, D. Kolarich, R. Strasser, and F. Altmann, Sialic acid concentrations in plants are in the range of inadvertent contamination, Planta, vol.224, pp.222-227, 2006.

T. Zhao, W. Wang, X. Bai, and Y. Qi, Gene silencing by artificial microRNAs in Chlamydomonas, Plant J, vol.58, pp.157-164, 2009.

S. Zhu, A. Hanneman, V. N. Reinhold, A. M. Spence, and H. Schachter, Caenorhabditis elegans triple null mutant lacking UDP-N-acetyl-D-glucosamine:?-3-D-mannoside ?1,2-N-acetylglucosaminyltransferase I, Biochem. J, vol.382, pp.995-1001, 2004.

D. F. Zielinska, F. Gnad, J. R. Wi?niewski, and M. Mann, Precision mapping of an in vivo N-glycoproteome reveals rigid topological and sequence constraints, Cell, vol.141, pp.897-907, 2010.

, Conflict of Interest Statement: The authors declare that the research was con

I. Chapitre, Comparative transcriptome analysis of P. tricornutum's morphotypes Chapitre III

A. S. Abdullahi, G. J. Underwood, and M. R. Gretz, Extracellular matrix assembly in diatoms (Bacillariophyceae). V. Environmental effects on polysaccharide synthesis in the model diatom, Phaeodactylum tricornutum, J. Phycol, vol.42, pp.363-378, 2006.

M. Aebi, N-linked protein glycosylation in the ER, Biochim. Biophys. Acta BBA -Mol. Cell Res, vol.1833, pp.2430-2437, 2013.

M. Aebi, J. Gassenhuber, H. Domdey, and S. Heesen, Cloning and characterization of the ALG3 gene of Saccharomyces cerevisiae, Glycobiology, vol.6, pp.439-444, 1996.

M. Aebi, R. Bernasconi, S. Clerc, and M. Molinari, N-glycan structures: recognition and processing in the ER, Trends Biochem. Sci, vol.35, pp.74-82, 2010.

J. Aikawa, Y. Takeda, I. Matsuo, and Y. Ito, Trimming of glucosylated N-glycans by human ER 1,2-mannosidase I, J. Biochem. (Tokyo), vol.155, pp.375-384, 2014.

F. Altmann and L. März, Processing of asparagine-linked oligosaccharides in insect cells: evidence for alpha-mannosidase II, Glycoconj. J, vol.12, pp.150-155, 1995.

S. Anders, P. T. Pyl, and W. Huber, HTSeq--a Python framework to work with highthroughput sequencing data, Bioinformatics, vol.31, pp.166-169, 2015.

C. L. Andersen, J. L. Jensen, and T. F. Ørntoft, Normalization of real-time quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets, Cancer Res, vol.64, pp.5245-5250, 2004.

K. E. Apt, A. R. Grossman, and P. G. Kroth-pancic, Stable nuclear transformation of the diatomPhaeodactylum tricornutum, Mol. Gen. Genet. MGG, vol.252, pp.572-579, 1996.

R. Apweiler, H. Hermjakob, S. , and N. , On the frequency of protein glycosylation, as deduced from analysis of the SWISS-PROT database, Biochim. Biophys. Acta BBA-Gen, 1999.

. Subj, , vol.1473, pp.4-8

E. V. Armbrust, The life of diatoms in the world's oceans, Nature, vol.459, pp.185-192, 2009.

E. V. Armbrust, J. A. Berges, C. Bowler, B. R. Green, D. Martinez et al., The genome of the diatom Thalassiosira pseudonana: ecology, evolution, and metabolism, Science, vol.306, pp.79-86, 2004.

M. Audry, C. Jeanneau, A. Imberty, A. Harduin-lepers, P. Delannoy et al., Current trends in the structure-activity relationships of sialyltransferases, Glycobiology, vol.21, pp.716-726, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01224155

B. Baiet, C. Burel, B. Saint-jean, R. Louvet, L. Menu-bouaouiche et al., N-Glycans of Phaeodactylum tricornutum Diatom and Functional Characterization of Its NAcetylglucosaminyltransferase I Enzyme, J. Biol. Chem, vol.286, pp.6152-6164, 2011.

H. Bakker, M. Bardor, J. W. Molthoff, V. Gomord, I. Elbers et al., Galactose-extended glycans of antibodies produced by transgenic plants, Proc. Natl. Acad. Sci, vol.98, pp.2899-2904, 2001.

S. L. Baldauf, A. J. Roger, I. Wenk-siefert, and W. F. Doolittle, A Kingdom-Level Phylogeny of Eukaryotes Based on Combined Protein Data, Science, vol.290, pp.972-977, 2000.

M. Bardor, C. Faveeuw, A. Fitchette, D. Gilbert, L. Galas et al., Immunoreactivity in mammals of two typical plant glyco-epitopes, core ? (1, 3)-fucose and core xylose, Glycobiology, vol.13, pp.427-434, 2003.

M. K. Barker and D. R. Rose, Specificity of Processing -Glucosidase I Is Guided by the Substrate Conformation: CRYSTALLOGRAPHIC AND IN SILICO STUDIES, J. Biol. Chem, vol.288, pp.13563-13574, 2013.

A. Bartual, J. A. Gálvez, and F. Ojeda, Phenotypic response of the diatom Phaeodactylum tricornutum Bohlin to experimental changes in the inorganic carbon system, 1951.

A. Bartual, B. Villazán, and F. G. Brun, Monitoring the long-term stability of pelagic morphotypes in the model diatom Phaeodactylum tricornutum, Diatom Res, vol.26, pp.243-253, 2011.

E. Bause, Structural requirements of N-glycosylation of proteins. Studies with proline peptides as conformational probes, Biochem. J, vol.209, pp.331-336, 1983.

E. W. Becker, Micro-algae as a source of protein, Biotechnol. Adv, vol.25, pp.207-210, 2007.

M. M. Van-beers and M. Bardor, Minimizing immunogenicity of biopharmaceuticals by controlling critical quality attributes of proteins, Biotechnol. J, vol.7, pp.1473-1484, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01844668

J. Bennett, D. Hondred, and J. C. Register, Keeping qRT-PCR rigorous and biologically relevant, Plant Cell Rep, vol.34, pp.1-3, 2015.

E. G. Berger and F. J. Hesford, Localization of galactosyl-and sialyltransferase by immunofluorescence: evidence for different sites, Proc. Natl. Acad. Sci, vol.82, pp.4736-4739, 1985.

T. Bickel, L. Lehle, M. Schwarz, M. Aebi, J. et al., Biosynthesis of lipidlinked oligosaccharides in Saccharomyces cerevisiae: Alg13p and Alg14p form a complex required for the formation of GlcNAc(2)-PP-dolichol, J. Biol. Chem, vol.280, pp.34500-34506, 2005.

J. Blackwell, K. D. Parker, and K. M. Rudall, Chitin fibres of the diatoms Thalassiosira fluviatilis and Cyclotella cryptica, J. Mol. Biol, vol.28, pp.383-385, 1967.

K. H. Bohlin, Zur Morphologie und Biologie einzelliger Algen, 1897.

M. A. Borowitzka and B. E. Volcani, The polymorphic diatom Phaeodactylum tricornutum: ultrastructure of its morphotypes, J. Phycol, vol.14, pp.10-21, 1978.

P. Both, L. Sobczak, C. Breton, S. Hann, K. Nobauer et al., Distantly related plant and nematode core 1,3-fucosyltransferases display similar trends in structure-function relationships, Glycobiology, vol.21, pp.1401-1415, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00757511

C. Bowler, A. E. Allen, J. H. Badger, J. Grimwood, K. Jabbari et al., The Phaeodactylum genome reveals the evolutionary history of diatom genomes, Nature, vol.456, pp.239-244, 2008.
URL : https://hal.archives-ouvertes.fr/cea-00910244

C. Bowler, A. Vardi, A. , and A. E. , Oceanographic and Biogeochemical Insights from Diatom Genomes, Annu. Rev. Mar. Sci, vol.2, pp.333-365, 2010.

A. Bozarth, U. Maier, and S. Zauner, Diatoms in biotechnology: modern tools and applications, Appl. Microbiol. Biotechnol, vol.82, pp.195-201, 2009.

D. Brada and U. C. Dubach, Isolation of a homogeneous glucosidase II from pig kidney microsomes, Eur. J. Biochem, vol.141, pp.149-156, 1984.

C. Breton, Structures and mechanisms of glycosyltransferases, Glycobiology, vol.16, pp.29-37, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00305830

C. Breton and A. Imberty, Structure/function studies of glycosyltransferases, Curr. Opin. Struct. Biol, vol.9, pp.563-571, 1999.

C. Breton, R. Oriol, and A. Imberty, Conserved structural features in eukaryotic and prokaryotic fucosyltransferases, Glycobiology, vol.8, pp.87-94, 1998.
URL : https://hal.archives-ouvertes.fr/hal-00302515

C. Breton, E. Bettler, D. H. Joziasse, R. A. Geremia, and A. Imberty, Sequencefunction relationships of prokaryotic and eukaryotic galactosyltransferases, J. Biochem. (Tokyo), vol.123, pp.1000-1009, 1998.
URL : https://hal.archives-ouvertes.fr/hal-00314475

W. Breuer, R. A. Klein, B. Hardt, A. Bartoschek, and E. Bause, , 2001.

, Oligosaccharyltransferase is highly specific for the hydroxy amino acid in Asn-Xaa-Thr/Ser, FEBS Lett, vol.501, pp.106-110

A. Brighouse, J. B. Dacks, and M. C. Field, Rab protein evolution and the history of the eukaryotic endomembrane system, Cell. Mol. Life Sci, vol.67, pp.3449-3465, 2010.

I. Brockhausen and H. Schachter, Glycosyltransferases Involved in N-and O-Glycan Biosynthesis, pp.79-113, 1996.

P. Burda, A. , and M. , The ALG10 locus of Saccharomyces cerevisiae encodes the alpha-1,2 glucosyltransferase of the endoplasmic reticulum: the terminal glucose of the lipidlinked oligosaccharide is required for efficient N-linked glycosylation, Glycobiology, vol.8, pp.455-462, 1998.

P. Burda, C. A. Jakob, J. Beinhauer, J. H. Hegemann, A. et al., Ordered assembly of the asymmetrically branched lipid-linked oligosaccharide in the endoplasmic reticulum is ensured by the substrate specificity of the individual glycosyltransferases, Glycobiology, vol.9, pp.617-625, 1999.

S. A. Bustin, Absolute quantification of mRNA using real-time reverse transcription polymerase chain reaction assays, J. Mol. Endocrinol, vol.25, pp.169-193, 2000.

S. A. Bustin, Why the need for qPCR publication guidelines?--The case for MIQE, Methods San Diego Calif, vol.50, pp.217-226, 2010.

J. J. Caramelo, O. A. Castro, L. G. Alonso, G. De-prat-gay, and A. J. Parodi, UDP-Glc: glycoprotein glucosyltransferase recognizes structured and solvent accessible hydrophobic patches in molten globule-like folding intermediates, Proc. Natl. Acad. Sci, vol.100, pp.86-91, 2003.

A. Castilho, M. Pabst, R. Leonard, C. Veit, F. Altmann et al., Construction of a functional CMP-sialic acid biosynthesis pathway in Arabidopsis, Plant Physiol, vol.147, pp.331-339, 2008.

A. Castilho, L. Neumann, P. Gattinger, R. Strasser, K. Vorauer-uhl et al., Generation of Biologically Active Multi-Sialylated Recombinant Human EPOFc in Plants, PLoS ONE, vol.8, p.54836, 2013.

A. Castilho, M. Windwarder, P. Gattinger, L. Mach, R. Strasser et al., Proteolytic and N-Glycan Processing of Human 1-Antitrypsin Expressed in Nicotiana benthamiana, Plant Physiol, vol.166, pp.1839-1851, 2014.

D. S. Chao, J. C. Hay, S. Winnick, R. Prekeris, J. Klumperman et al., SNARE membrane trafficking dynamics in vivo, J. Cell Biol, vol.144, pp.869-881, 1999.

J. R. Chapman and J. Waldenström, With Reference to Reference Genes: A Systematic Review of Endogenous Controls in Gene Expression Studies, PloS One, vol.10, 2015.

M. M. Chen, K. J. Glover, and B. Imperiali, From Peptide to Protein: Comparative Analysis of the Substrate Specificity of N-Linked Glycosylation in C. jejuni ?, Biochemistry (Mosc.), vol.46, pp.5579-5585, 2007.

F. Y. Cheong, V. Sharma, A. Blagoveshchenskaya, V. M. Oorschot, B. Brankatschk et al., Spatial Regulation of Golgi Phosphatidylinositol-4-Phosphate is Required for Enzyme Localization and Glycosylation Fidelity, Traffic, vol.11, pp.1180-1190, 2010.

V. A. Chepurnov, D. G. Mann, K. Sabbe, and W. Vyverman, Experimental Studies on Sexual Reproduction in Diatoms, International Review of Cytology, pp.91-154, 2004.

L. Chevalier, S. Bernard, Y. Ramdani, R. Lamour, M. Bardor et al., Subcompartment localization of the side chain xyloglucansynthesizing enzymes within Golgi stacks of tobacco suspension-cultured cells: Golgi localization of xyloglucan-synthesizing enzymes, Plant J, vol.64, pp.977-989, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01848389

A. Chiovitti, R. E. Harper, A. Willis, A. Bacic, P. Mulvaney et al., , 2005.

, Variations in the substituted 3-linked mannans closely associated with the silicified walls of diatoms : substituted mannans of diatoms, J. Phycol, vol.41, pp.1154-1161

Y. H. Choi, J. H. Kim, J. H. Park, N. Lee, D. Kim et al., Protein engineering of 2,3/2,6-sialyltransferase to improve the yield and productivity of in vitro sialyllactose synthesis, Glycobiology, vol.24, pp.159-169, 2014.

L. L. Christensen, U. B. Jensen, P. Bross, and T. F. Orntoft, The C-terminal Nglycosylation sites of the human alpha1,3/4-fucosyltransferase III, -V, and -VI (hFucTIII, -V, adn -VI) are necessary for the expression of full enzyme activity, Glycobiology, vol.10, pp.931-939, 2000.

A. Ciechanover, A. Orian, and A. L. Schwartz, Ubiquitin-mediated proteolysis: biological regulation via destruction, BioEssays, vol.22, pp.442-451, 2000.

J. F. Cipollo, R. B. Trimble, J. H. Chi, Q. Yan, and N. Dean, The Yeast ALG11 Gene Specifies Addition of the Terminal 1,2-Man to the, 2001.

, Références bibliographiques

, Intermediate Formed on the Cytosolic Side of the Endoplasmic Reticulum, J. Biol. Chem, vol.276, pp.21828-21840

R. A. Clynes, T. L. Towers, L. G. Presta, and J. V. Ravetch, Inhibitory Fc receptors modulate in vivo cytoxicity against tumor targets, Nat. Med, vol.6, pp.443-446, 2000.

S. Coesel, M. Oborník, J. Varela, A. Falciatore, and C. Bowler, Evolutionary Origins and Functions of the Carotenoid Biosynthetic Pathway in Marine Diatoms, PLoS ONE, vol.3, 2008.

A. Conesa, S. Gotz, J. M. Garcia-gomez, J. Terol, M. Talon et al., , 2005.

, Blast2GO: a universal tool for annotation, visualization and analysis in functional genomics research, Bioinformatics, vol.21, pp.3674-3676

A. Conesa, P. Madrigal, S. Tarazona, D. Gomez-cabrero, A. Cervera et al., A survey of best practices for RNA-seq data analysis, Genome Biol, vol.17, 2016.

B. J. Conti, P. K. Devaraneni, Z. Yang, L. L. David, and W. R. Skach, Cotranslational Stabilization of Sec62/63 within the ER Sec61 Translocon Is Controlled by Distinct SubstrateDriven Translocation Events, Mol. Cell, vol.58, pp.269-283, 2015.

K. E. Cooksey and B. Cooksey, Calcium deficiency can induce the transition from oval to fusiform cells in cultures of Phaeodactylum tricornutum Bohlin, J. Phycol, vol.10, pp.89-90, 1974.

J. R. Couto, T. C. Huffaker, and P. W. Robbins, Cloning and expression in Escherichia coli of a yeast mannosyltransferase from the asparagine-linked glycosylation pathway, J. Biol. Chem, vol.259, pp.378-382, 1984.

E. K. Culyba, J. L. Price, S. R. Hanson, A. Dhar, C. Wong et al., Protein Native-State Stabilization by Placing Aromatic Side Chains in N-Glycosylated Reverse Turns, Science, vol.331, pp.571-575, 2011.

T. Czechowski, M. Stitt, T. Altmann, M. K. Udvardi, and W. Scheible, Genomewide identification and testing of superior reference genes for transcript normalization in Arabidopsis, Plant Physiol, vol.139, pp.5-17, 2005.

J. Czlapinski and C. Bertozzi, Synthetic glycobiology: exploits in the Golgi compartment, Curr. Opin. Chem. Biol, vol.10, pp.645-651, 2006.

W. M. Darley, Deoxyribonucleic acid content of the three cell types of Phaeodactylum tricornutum Bohlin, J. Phycol, vol.4, pp.219-220, 1968.

D. Martino, A. Meichenin, A. Shi, J. Pan, K. Bowler et al., Genetic and phenotypic characterization of Phaeodactylum tricornutum (Bacillariophyceae) accessions, J. Phycol, vol.43, pp.992-1009, 2007.

D. Martino, A. Bartual, A. Willis, A. Meichenin, A. Villazán et al., Physiological and Molecular Evidence that Environmental Changes Elicit Morphological Interconversion in the Model Diatom Phaeodactylum tricornutum, Protist, vol.162, pp.462-481, 2011.

K. De-pourcq, K. De-schutter, and N. Callewaert, Engineering of glycosylation in yeast and other fungi: current state and perspectives, Appl. Microbiol. Biotechnol, vol.87, pp.1617-1631, 2010.

V. De-riso, R. Raniello, F. Maumus, A. Rogato, C. Bowler et al., Gene silencing in the marine diatom Phaeodactylum tricornutum, Nucleic Acids Res, vol.37, pp.96-96, 2009.

S. Y. Dejgaard, A. Murshid, K. M. Dee, and J. F. Presley, Confocal microscopy-based linescan methodologies for intra-Golgi localization of proteins, J. Histochem. Cytochem, vol.55, pp.709-719, 2007.

P. Deprez, M. Gautschi, and A. Helenius, More Than One Glycan Is Needed for ER Glucosidase II to Allow Entry of Glycoproteins into the Calnexin/Calreticulin Cycle, Mol. Cell, vol.19, pp.183-195, 2005.

E. L. Van-dijk, Y. Jaszczyszyn, and C. Thermes, Library preparation methods for nextgeneration sequencing: tone down the bias, Exp. Cell Res, vol.322, pp.12-20, 2014.

R. Dillschneider, C. Steinweg, R. Rosello-sastre, and C. Posten, Biofuels from microalgae: Photoconversion efficiency during lipid accumulation, Bioresour. Technol, vol.142, pp.647-654, 2013.

B. S. Donohoe, B. Kang, M. J. Gerl, Z. R. Gergely, C. M. Mcmichael et al., Cis -Golgi Cisternal Assembly and Biosynthetic Activation Occur Sequentially in Plants and Algae: Cis-Golgi Cisternal Assembly and Biosynthetic Activation in Plants and Algae, Traffic, vol.14, pp.551-567, 2013.

T. M. Dugdale, A. Willis, and R. Wetherbee, Adhesive Modular Proteins Occur in the Extracellular Mucilage of the Motile, Pennate Diatom Phaeodactylum tricornutum, Biophys. J, vol.90, pp.58-60, 2006.

F. Dupuy, J. Petit, R. Mollicone, R. Oriol, R. Julien et al., A Single Amino Acid in the Hypervariable Stem Domain of Vertebrate ?1, 3/1, 4-Fucosyltransferases Determines the Type 1/Type 2 Transfer CHARACTERIZATION OF ACCEPTOR SUBSTRATE SPECIFICITY OF THE LEWIS ENZYME BY SITE-DIRECTED MUTAGENESIS, J. Biol. Chem, vol.274, pp.12257-12262, 1999.
URL : https://hal.archives-ouvertes.fr/hal-01211866

E. Eisenberg and E. Y. Levanon, Human housekeeping genes, revisited, Trends Genet. TIG, vol.29, pp.569-574, 2013.

L. Ellgaard and A. Helenius, ER quality control: towards an understanding at the molecular level, Curr. Opin. Cell Biol, vol.13, pp.431-437, 2001.

L. Ellgaard and A. Helenius, Quality control in the endoplasmic reticulum, Nat. Rev. Mol. Cell Biol, vol.4, pp.181-191, 2003.

G. Fabini, A. Freilinger, F. Altmann, W. , and I. , Identification of Core ?1,3-Fucosylated Glycans and Cloning of the Requisite Fucosyltransferase cDNA from Drosophila melanogaster, 2001.

A. Falciatore, R. Casotti, C. Leblanc, C. Abrescia, and C. Bowler, Transformation of nonselectable reporter genes in marine diatoms, Mar. Biotechnol, vol.1, pp.239-251, 1999.

L. Faye, A. Sturm, R. Bollini, A. Vitale, and M. J. Chrispeels, The position of the oligosaccharide side-chains of phytohemagglutinin and their accessibility to glycosidases determines their subsequent processing in the Golgi, Eur. J. Biochem, vol.158, pp.655-661, 1986.

K. Fiedler, R. G. Parton, R. Kellner, T. Etzold, and K. Simons, VIP36, a novel component of glycolipid rafts and exocytic carrier vesicles in epithelial cells, EMBO J, vol.13, p.1729, 1994.

C. B. Field, M. J. Behrenfeld, J. T. Randerson, and P. Falkowski, Primary production of the biosphere: integrating terrestrial and oceanic components, Science, vol.281, pp.237-240, 1998.

A. C. Fitchette, M. Cabanes-macheteau, L. Marvin, B. Martin, B. Satiat-jeunemaitre et al., Biosynthesis and immunolocalization of Lewis a-containing N-glycans in the plant cell, Plant Physiol, vol.121, pp.333-344, 1999.

A. Fitchette-lainé, V. Gomord, A. Chekkafi, F. , and L. , Distribution of xylosylation and fucosylation in the plant Golgi apparatus, Plant J, vol.5, pp.673-682, 1994.

A. C. Fitchette-lainé, V. Gomord, M. Cabanes, J. C. Michalski, M. Saint-macary et al., N-glycans harboring the Lewis a epitope are expressed at the surface of plant cells, Plant J. Cell Mol. Biol, vol.12, pp.1411-1417, 1997.

C. W. Ford, P. , and E. , The carbohydrates of Phaeodactylum tricornutum. Part I. Preliminary examination of the organism, and characterisation of low molecular weight material and of a glucan, J. Chem. Soc. Resumed, pp.7035-7041, 1965.

G. Francius, B. Tesson, E. Dague, V. Martin-jézéquel, and Y. F. Dufrêne, , 2008.

, Nanostructure and nanomechanics of live Phaeodactylum tricornutum morphotypes, Environ. Microbiol, vol.10, pp.1344-1356

C. G. Frank, A. , and M. , ALG9 mannosyltransferase is involved in two different steps of lipid-linked oligosaccharide biosynthesis, Glycobiology, vol.15, pp.1156-1163, 2005.

E. Frickel, R. Riek, I. Jelesarov, A. Helenius, K. Wüthrich et al., , 2002.

, TROSY-NMR reveals interaction between ERp57 and the tip of the calreticulin P-domain

, Proc. Natl. Acad. Sci, vol.99, pp.1954-1959

T. Fukuda, H. Hashimoto, N. Okayasu, A. Kameyama, H. Onogi et al., 1,6-Fucosyltransferase-deficient Mice Exhibit Multiple Behavioral Abnormalities Associated with a Schizophrenia-like Phenotype: IMPORTANCE OF THE BALANCE BETWEEN THE DOPAMINE AND SEROTONIN SYSTEMS, J. Biol. Chem, vol.286, pp.18434-18443, 2011.

C. Gachon, A. Mingam, and B. Charrier, Real-time PCR: what relevance to plant studies, J. Exp. Bot, vol.55, pp.1445-1454, 2004.

X. Gao, A. Nishikawa, and N. Dean, Physical interactions between the Alg1, Alg2, and Alg11 mannosyltransferases of the endoplasmic reticulum, Glycobiology, vol.14, pp.559-570, 2004.

Y. Gavel and G. Heijne, Sequence differences between glycosylated and nonglycosylated Asn-X-Thr/Ser acceptor sites: implications for protein engineering, Protein Eng, vol.3, pp.433-442, 1990.

J. Gillard, J. Frenkel, V. Devos, K. Sabbe, C. Paul et al., Metabolomics Enables the Structure Elucidation of a Diatom Sex Pheromone, Angew. Chem. Int. Ed, vol.52, pp.854-857, 2013.

M. H. Glickman and A. Ciechanover, The Ubiquitin-Proteasome Proteolytic Pathway: Destruction for the Sake of Construction, Physiol. Rev, vol.82, pp.373-428, 2002.

S. Guénin, M. Mauriat, J. Pelloux, O. Van-wuytswinkel, C. Bellini et al., Normalization of qRT-PCR data: the necessity of adopting a systematic, experimental conditions-specific, validation of references, J. Exp. Bot, vol.60, pp.487-493, 2009.

M. Guerin and A. J. Parodi, The UDP-glucose:Glycoprotein Glucosyltransferase Is Organized in at Least Two Tightly Bound Domains from Yeast to Mammals, J. Biol. Chem, vol.278, pp.20540-20546, 2003.

B. Gügi, T. Costaouec, C. Burel, P. Lerouge, W. Helbert et al., DiatomSpecific Oligosaccharide and Polysaccharide Structures Help to Unravel Biosynthetic Capabilities in Diatoms, Mar. Drugs, vol.13, pp.5993-6018, 2015.

S. A. Gutenbrunner, J. Thalhamer, and A. M. Schmid, Proteinaceous and immunochemical distinctions between the oval and fusiform morphotypes of Phaeodactylum tricornutum (Bacillariophyceae), J. Phycol, vol.30, pp.129-136, 1994.

L. Gutierrez, M. Mauriat, J. Pelloux, C. Bellini, and O. Van-wuytswinkel, Towards a systematic validation of references in real-time rt-PCR, Plant Cell, vol.20, pp.1734-1735, 2008.

M. Hahn, S. Borisova, J. D. Schrag, D. C. Tessier, A. Zapun et al., Identification and Crystallization of a Protease-Resistant Core of Calnexin That Retains Biological Activity, J. Struct. Biol, vol.123, pp.260-264, 1998.

S. R. Hanson, E. K. Culyba, T. Hsu, C. Wong, J. W. Kelly et al., The core trisaccharide of an N-linked glycoprotein intrinsically accelerates folding and enhances stability, Proc. Natl. Acad. Sci, vol.106, pp.3131-3136, 2009.

G. R. Hasle, The Marine, Planktonic Diatoms Thalassiosira-Oceanica Sp-Nov and Thalassiosira-Partheneia, J. Phycol, vol.19, pp.220-229, 1983.

H. Hauri, F. Kappeler, H. Andersson, and C. Appenzeller, ERGIC-53 and traffic in the secretory pathway, J. Cell Sci, vol.113, pp.587-596, 2000.

L. He, X. Han, Y. , and Z. , A Rare Phaeodactylum tricornutum Cruciform Morphotype: Culture Conditions, Transformation and Unique Fatty Acid Characteristics, PLoS ONE, vol.9, p.93922, 2014.

R. E. Hecky, K. Mopper, P. Kilham, and E. T. Degens, The amino acid and sugar composition of diatom cell-walls, Mar. Biol, vol.19, pp.323-331, 1973.

S. Heesen, L. Lehle, A. Weissmann, A. , and M. , Isolation of the ALG5 Locus Encoding the UDP-Glucose: Dolichyl-Phosphate Glucosyltransferase from Saccharomyces cerevisiae, Eur. J. Biochem, vol.224, pp.71-79, 1994.

A. Helenius, How N-linked oligosaccharides affect glycoprotein folding in the endoplasmic reticulum, Mol. Biol. Cell, vol.5, p.253, 1994.

A. Helenius, A. , and M. , Roles of N-linked glycans in the endoplasmic reticulum, Annu. Rev. Biochem, vol.73, pp.1019-1049, 2004.

J. Helenius, D. T. Ng, C. L. Marolda, P. Walter, M. A. Valvano et al., , 2002.

, Translocation of lipid-linked oligosaccharides across the ER membrane requires Rft1 protein, Nature, vol.415, pp.447-450

W. Hemmer, M. Focke, D. Kolarich, I. B. Wilson, F. Altmann et al., Antibody binding to venom carbohydrates is a frequent cause for double positivity to honeybee and yellow jacket venom in patients with stinging-insect allergy, J. Allergy Clin. Immunol, vol.108, pp.1045-1052, 2001.

F. Hempel, J. Lau, A. Klingl, and U. G. Maier, Algae as Protein Factories: Expression of a Human Antibody and the Respective Antigen in the Diatom Phaeodactylum tricornutum, PLoS ONE, vol.6, 2011.

F. Hempel, A. S. Bozarth, N. Lindenkamp, A. Klingl, S. Zauner et al., Microalgae as bioreactors for bioplastic production, Microb Cell Fact, vol.10, p.81, 2011.

N. I. Hendey, D. H. Cushing, and G. W. Ripley, Electron microscope studies of diatoms, J. R. Microsc. Soc, vol.74, pp.22-34, 1954.

H. Hettkamp, G. Legler, and E. Bause, Purification by affinity chromatography of glucosidase I, an endoplasmic reticulum hydrolase involved in the processing of asparaginelinked oligosaccharides, Eur. J. Biochem, vol.142, pp.85-90, 1984.

M. J. Higgins, P. Molino, P. Mulvaney, and R. Wetherbee, The structure and nanomechanical properties of the adhesive mucilage that mediates diatom-substratum adhesion and motility, J. Phycol, vol.39, pp.1181-1193, 2003.

S. High, F. J. Lecomte, S. J. Russell, B. M. Abell, and J. D. Oliver, Glycoprotein folding in the endoplasmic reticulum: a tale of three chaperones?, FEBS Lett, vol.476, pp.38-41, 2000.

M. Hildebrand, B. E. Volcani, W. Gassmann, and J. I. Schroeder, A gene family of silicon transporters, Nature, vol.385, pp.688-689, 1997.

M. Hildebrand, K. Dahlin, and B. E. Volcani, Characterization of a silicon transporter gene family in Cylindrotheca fusiformis: sequences, expression analysis, and identification of homologs in other diatoms, Mol. Gen. Genet. MGG, vol.260, pp.480-486, 1998.

C. Hoek, . Van-den, D. G. Mann, and H. M. Jahns, Algae: an introduction to phycology (Cambridge, 1995.

E. H. Holmes, T. Yen, S. Thomas, R. Joshi, A. Nguyen et al., Human ?1, 3/4 Fucosyltransferases CHARACTERIZATION OF HIGHLY CONSERVED CYSTEINE RESIDUES AND N-LINKED GLYCOSYLATION SITES, J. Biol. Chem, vol.275, pp.24237-24245, 2000.
URL : https://hal.archives-ouvertes.fr/hal-01211856

S. Holst, A. J. Deuss, G. W. Van-pelt, S. J. Van-vliet, J. J. Garcia-vallejo et al., Nglycosylation Profiling of Colorectal Cancer Cell Lines Reveals Association of Fucosylation with Differentiation and Caudal Type Homebox 1 (CDX1)/Villin mRNA Expression, Mol. Cell. Proteomics, vol.15, pp.124-140, 2016.

T. Hruz, M. Wyss, M. Docquier, M. W. Pfaffl, S. Masanetz et al., RefGenes: identification of reliable and condition specific reference genes for RT-qPCR data normalization, BMC Genomics, vol.12, p.156, 2011.

A. Huang, L. He, W. , and G. , Identification and characterization of microRNAs from Phaeodactylum tricornutum by high-throughput sequencing and bioinformatics analysis, BMC Genomics, vol.12, 2011.

J. Huggett, K. Dheda, S. Bustin, and A. Zumla, Real-time RT-PCR normalisation; strategies and considerations, Genes Immun, vol.6, pp.279-284, 2005.

M. J. Huysman, C. Martens, K. Vandepoele, J. Gillard, E. Rayko et al., Genome-wide analysis of the diatom cell cycle unveils a novel type of cyclins involved in environmental signaling, Genome Biol, vol.11, p.17, 2010.

S. Iida, Nonfucosylated Therapeutic IgG1 Antibody Can Evade the Inhibitory Effect of Serum Immunoglobulin G on Antibody-Dependent Cellular Cytotoxicity through its High Binding to Fc RIIIa, Clin. Cancer Res, vol.12, pp.2879-2887, 2006.

A. Imberty, C. Monier, E. Bettler, S. Morera, P. Freemont et al., Fold recognition study of ?3-galactosyltransferase and molecular modeling of the nucleotide sugar-binding domain, Glycobiology, vol.9, pp.713-722, 1999.
URL : https://hal.archives-ouvertes.fr/hal-00309797

B. Imperiali and T. L. Hendrickson, Asparagine-linked glycosylation: Specificity and function of oligosaccharyl transferase, Bioorg. Med. Chem, vol.3, pp.1565-1578, 1995.

B. Imperiali and S. E. Connor, Effect of N-linked glycosylation on glycopeptide and glycoprotein structure, Curr. Opin. Chem. Biol, vol.3, pp.643-649, 1999.

C. Jeanneau, Structure-Function Analysis of the Human Sialyltransferase ST3Gal I: ROLE OF N-GLYCOSYLATION AND A NOVEL CONSERVED SIALYL MOTIF, J. Biol. Chem, vol.279, pp.13461-13468, 2004.

R. Jefferis, J. Lund, and J. D. Pound, IgG-Fc-mediated effector functions: molecular definition of interaction sites for effector ligands and the role of glycosylation, Immunol. Rev, vol.163, pp.59-76, 1998.

K. G. Jensen, Ø. Moestrup, and A. M. Schmid, Ultrastructure of the male gametes from two centric diatoms, Chaetoceros laciniosus and Coscinodiscus wailesii (Bacillariophyceae), Phycologia, vol.42, pp.98-105, 2003.

J. Jez, A. Castilho, J. Grass, K. Vorauer-uhl, T. Sterovsky et al., Expression of functionally active sialylated human erythropoietin in plants, Biotechnol. J, vol.8, pp.371-382, 2013.

J. R. Johansen, Morphological variability and cell wall composition of Phaeodactylum tricornutum (Bacillariophyceae), Gt. Basin Nat, vol.51, pp.310-315, 1991.

K. D. Johnson and M. J. Chrispeels, Substrate specificities of N-acetylglucosaminyl-, fucosyl-, and xylosyltransferases that modify glycoproteins in the Golgi apparatus of bean cotyledons, Plant Physiol, vol.84, pp.1301-1308, 1987.

H. J. De-jonge, R. S. Fehrmann, E. S. De-bont, R. M. Hofstra, F. Gerbens et al., Evidence based selection of housekeeping genes, PloS One, vol.2, 2007.

F. Jost, Mutation of amino acids in the alpha 1,3-fucosyltransferase motif affects enzyme activity and Km for donor and acceptor substrates, Glycobiology, vol.15, pp.165-175, 2004.

J. Kaewmuangmoon, M. Kilaso, U. Leartsakulpanich, K. Kimura, A. Kimura et al., Expression of a secretory ?-glucosidase II from Apis cerana indica in Pichia pastoris and its characterization, BMC Biotechnol, vol.13, p.16, 2013.

S. Kallolimath, A. Castilho, R. Strasser, C. Grünwald-gruber, F. Altmann et al., Engineering of complex protein sialylation in plants, Proc. Natl. Acad. Sci, vol.113, pp.9498-9503, 2016.

M. Kampf, B. Absmanner, M. Schwarz, and L. Lehle, Biochemical Characterization and Membrane Topology of Alg2 from Saccharomyces cerevisiae as a Bifunctional 1,3-and 1,6-Mannosyltransferase Involved in Lipid-linked Oligosaccharide Biosynthesis, J. Biol. Chem, vol.284, pp.11900-11912, 2009.

B. J. Karas, R. E. Diner, S. C. Lefebvre, J. Mcquaid, A. P. Phillips et al., Designer diatom episomes delivered by bacterial conjugation, Nat. Commun, vol.6, p.6925, 2015.

G. P. Kaushal, T. Szumilo, I. Pastuszak, and A. D. Elbein, Purification to homogeneity and properties of mannosidase II from mung bean seedlings, Biochemistry (Mosc.), vol.29, pp.2168-2176, 1990.

P. J. Keeling, The Number, Speed, and Impact of Plastid Endosymbioses in Eukaryotic Evolution, Annu. Rev. Plant Biol, vol.64, pp.583-607, 2013.

P. J. Keeling and J. D. Palmer, Horizontal gene transfer in eukaryotic evolution, Nat. Rev. Genet, vol.9, pp.605-618, 2008.

D. J. Kelleher, S. Banerjee, A. J. Cura, J. Samuelson, and R. Gilmore, Dolichol-linked oligosaccharide selection by the oligosaccharyltransferase in protist and fungal organisms, J. Cell Biol, vol.177, pp.29-37, 2007.

G. Kern, D. Kern, R. Jaenicke, and R. Seckler, Kinetics of folding and association of differently glycosylated variants of invertase from Saccharomyces cerevisiae, Protein Sci, vol.2, pp.1862-1868, 1993.

D. Kim, G. Pertea, C. Trapnell, H. Pimentel, R. Kelley et al., TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions, 2013.

, Genome Biol, vol.14, p.1

A. Koprivova, F. Altmann, G. Gorr, S. Kopriva, R. Reski et al., NGlycosylation in the Moss Physcomitrella patens is Organized Similarly to that in Higher Plants, Plant Biol, vol.5, pp.582-591, 2003.

B. Kozera and M. Rapacz, Reference genes in real-time PCR, J. Appl. Genet, vol.54, pp.391-406, 2013.

G. Kozlov, P. Maattanen, J. D. Schrag, S. Pollock, M. Cygler et al., Crystal Structure of the bb? Domains of the Protein Disulfide Isomerase ERp57, Structure, vol.14, pp.1331-1339, 2006.

N. Kröger, Prescribing diatom morphology: toward genetic engineering of biological nanomaterials, Curr. Opin. Chem. Biol, vol.11, pp.662-669, 2007.

N. Kröger and N. Poulsen, Diatoms-From Cell Wall Biogenesis to Nanotechnology, Annu. Rev. Genet, vol.42, pp.83-107, 2008.

N. Kröger and R. Wetherbee, Pleuralins are Involved in Theca Differentiation in the Diatom Cylindrotheca fusiformis, Protist, vol.151, pp.263-273, 2000.

N. Kröger, R. Deutzmann, C. Bergsdorf, and M. Sumper, Species-specific polyamines from diatoms control silica morphology, Proc. Natl. Acad. Sci, vol.97, pp.14133-14138, 2000.

V. Kubelka, F. Altmann, E. Staudacher, V. Tretter, L. März et al., Primary structures of the N-linked carbohydrate chains from honeybee venom phospholipase A2, Eur. J. Biochem, vol.213, pp.1193-1204, 1993.

M. Kubista, J. M. Andrade, M. Bengtsson, A. Forootan, J. Jonák et al., The real-time polymerase chain reaction, Mol. Aspects Med, vol.27, pp.95-125, 2006.

P. Kuczynska, M. Jemiola-rzeminska, and K. Strzalka, Photosynthetic Pigments in Diatoms, Mar. Drugs, vol.13, pp.5847-5881, 2015.

I. Kudo, M. Miyamoto, Y. Noiri, and Y. Maita, Combined effects of temperature and iron on the growth and physiology of the marine diatom Phaeodactylum tricornutum (Bacillariophyceae), J. Phycol, vol.36, pp.1096-1102, 2000.

R. Kuliawat, E. Kalinina, J. Bock, L. Fricker, T. E. Mcgraw et al., Syntaxin-6 SNARE involvement in secretory and endocytic pathways of cultured pancreatic beta-cells, Mol. Biol. Cell, vol.15, pp.1690-1701, 2004.

S. Kurz, J. G. King, R. R. Dinglasan, K. Paschinger, and I. B. Wilson, The fucomic potential of mosquitoes: Fucosylated N-glycan epitopes and their cognate fucosyltransferases, Insect Biochem. Mol. Biol, vol.68, pp.52-63, 2016.

L. Lamriben, J. B. Graham, B. M. Adams, and D. N. Hebert, N-glycan based ER molecular chaperone and protein quality control system: the calnexin binding cycle, 2015.

B. Langmead and S. L. Salzberg, Fast gapped-read alignment with Bowtie 2, Nat. Methods, vol.9, pp.357-359, 2012.

B. Langmead, C. Trapnell, M. Pop, and S. L. Salzberg, Ultrafast and memory-efficient alignment of short DNA sequences to the human genome, Genome Biol, vol.10, p.25, 2009.

G. Lauc, ?. Kriå¡tiä, J. Zoldoå¡, and V. , Glycans -The third revolution in evolution, Front. Genet, vol.5, 2014.

R. E. Lee, Phycology, 1999.

R. Léonard, S. Lhernould, M. Carlué, P. Fleurat, A. Maftah et al., Biochemical characterization of Silene alba ?4-fucosyltransferase and Lewis a products, Glycoconj. J, vol.22, pp.71-78, 2005.

P. Lerouge, M. Cabanes-macheteau, C. Rayon, A. Fischette-lainé, V. Gomord et al., N-glycoprotein biosynthesis in plants: recent developments and future trends, Protein Trafficking in Plant Cells, pp.31-48, 1998.

J. C. Lewin, The taxonomic position of Phaeodactylum tricornutum, J. Gen. Microbiol, vol.18, pp.427-432, 1958.

J. C. Lewin, R. A. Lewin, and D. E. Philpott, Observations on Phaeodactylum tricornutum, J. Gen. Microbiol, vol.18, pp.418-426, 1958.

G. D. Lewis, I. Figari, B. Fendly, W. L. Wong, P. Carter et al., Differential responses of human tumor cell lines to anti-p185HER2 monoclonal antibodies, Cancer Immunol. Immunother, vol.37, pp.255-263, 1993.

M. J. Lewis, J. C. Rayner, and H. R. Pelham, A novel SNARE complex implicated in vesicle fusion with the endoplasmic reticulum, EMBO J, vol.16, pp.3017-3024, 1997.

Y. Li, X. Lei, H. Zhu, H. Zhang, C. Guan et al., Chitinase producing bacteria with direct algicidal activity on marine diatoms, Sci. Rep, vol.6, p.21984, 2016.

Z. Li, W. F. Stafford, and M. Bouvier, The Metal Ion Binding Properties of Calreticulin Modulate Its Conformational Flexibility and Thermal Stability ?, Biochemistry (Mosc.), vol.40, pp.11193-11201, 2001.

E. Liebminger, C. Veit, M. Pabst, M. Batoux, C. Zipfel et al., N-Acetylhexosaminidases HEXO1 and HEXO3 Are Responsible for the Formation of Paucimannosidic N-Glycans in Arabidopsis thaliana, J. Biol. Chem, vol.286, pp.10793-10802, 2011.

Y. Lin, C. Zhang, H. Lan, S. Gao, H. Liu et al., Validation of potential reference genes for qPCR in maize across abiotic stresses, hormone treatments, and tissue types, PloS One, vol.9, p.95445, 2014.

N. Lingg, P. Zhang, Z. Song, and M. Bardor, The sweet tooth of biopharmaceuticals: Importance of recombinant protein glycosylation analysis, Biotechnol. J, vol.7, pp.1462-1472, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01844671

Y. Liu and C. Barlowe, Analysis of Sec22p in endoplasmic reticulum/Golgi transport reveals cellular redundancy in SNARE protein function, Mol. Biol. Cell, vol.13, pp.3314-3324, 2002.

K. J. Livak and T. D. Schmittgen, Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2???CT Method, Methods, vol.25, pp.402-408, 2001.

C. Lizak, S. Gerber, S. Numao, M. Aebi, and K. P. Locher, X-ray structure of a bacterial oligosaccharyltransferase, Nature, vol.474, pp.350-355, 2011.

M. I. Love, W. Huber, A. , and S. , Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biol, vol.15, 2014.

Y. Lu and F. Wold, Regulation of glycan processing by Golgi enzymes from red kidney bean (Phaseolus vulgaris) seedlings, Arch. Biochem. Biophys, vol.286, pp.147-152, 1991.

J. Lu, T. Takahashi, A. Ohoka, K. Nakajima, R. Hashimoto et al., Alg14 organizes the formation of a multiglycosyltransferase complex involved in initiation of lipid-linked oligosaccharide biosynthesis, Glycobiology, vol.22, pp.504-516, 2012.

W. Luo and C. Brouwer, Pathview: an R/Bioconductor package for pathway-based data integration and visualization, Bioinformatics, vol.29, pp.1830-1831, 2013.

P. Määttänen, K. Gehring, J. J. Bergeron, and D. Y. Thomas, Protein quality control in the ER: The recognition of misfolded proteins, Semin. Cell Dev. Biol, vol.21, pp.500-511, 2010.

U. Maheswari, T. Mock, E. V. Armbrust, and C. Bowler, Update of the Diatom EST Database: a new tool for digital transcriptomics, Nucleic Acids Res, vol.37, pp.1001-1005, 2009.

U. Maheswari, K. Jabbari, J. Petit, B. M. Porcel, A. E. Allen et al., Digital expression profiling of novel diatom transcripts provides insight into their biological functions, Genome Biol, vol.11, p.85, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00627911

F. Mallard, B. L. Tang, T. Galli, D. Tenza, A. Saint-pol et al., Early/recycling endosomes-to-TGN transport involves two SNARE complexes and a Rab6 isoform, J. Cell Biol, vol.156, pp.653-664, 2002.

L. Malphettes, Y. Freyvert, J. Chang, P. Liu, E. Chan et al., Highly efficient deletion of FUT8 in CHO cell lines using zinc-finger nucleases yields cells that produce completely nonfucosylated antibodies, Biotechnol. Bioeng, vol.106, pp.774-783, 2010.

D. G. Mann, Patterns of sexual reproduction in diatoms, Twelfth International Diatom Symposium, pp.11-20, 1993.

D. G. Mann, The species concept in diatoms, Phycologia, vol.38, pp.437-495, 1999.

D. G. Mann and P. Vanormelingen, An Inordinate Fondness? The Number, Distributions, and Origins of Diatom Species, J. Eukaryot. Microbiol, vol.60, pp.414-420, 2013.

, Références bibliographiques

I. Manton, Observations on the fine structure of the male gamete of the marine centric diatom Lithodesmium undulatum, J. R. Microsc. Soc, vol.85, pp.119-134, 1966.

K. W. Marek, I. K. Vijay, M. , and J. D. , A recessive deletion in the GlcNAc-1-phosphotransferase gene results in peri-implantation embryonic lethality, Glycobiology, vol.9, pp.1263-1271, 1999.

V. Martin-jezequel, M. Hildebrand, and M. A. Brzezinski, Silicon Metabolism in Diatoms: Implications for Growth, 2000.

, J. Phycol, vol.36, pp.821-840

S. W. Mast and K. W. Moremen, Family 47 ?-Mannosidases in N-Glycan Processing, In Methods in Enzymology, pp.31-46, 2006.

K. Masuda, T. Kubota, E. Kaneko, S. Iida, M. Wakitani et al., Enhanced binding affinity for Fc?RIIIa of fucosenegative antibody is sufficient to induce maximal antibody-dependent cellular cytotoxicity, Mol. Immunol, vol.44, pp.3122-3131, 2007.

T. M. Mata, A. A. Martins, and N. S. Caetano, Microalgae for biodiesel production and other applications: A review, Renew. Sustain. Energy Rev, vol.14, pp.217-232, 2010.

E. Mathieu-rivet, M. Scholz, C. Arias, F. Dardelle, S. Schulze et al., Exploring the Nglycosylation Pathway in Chlamydomonas reinhardtii Unravels Novel Complex Structures, Mol. Cell. Proteomics, vol.12, pp.3160-3183, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00996460

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, Front. Plant Sci, vol.5, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01842175

F. Maumus, A. E. Allen, C. Mhiri, H. Hu, K. Jabbari et al., Potential impact of stress activated retrotransposons on genome evolution in a marine diatom, BMC Genomics, vol.10, p.624, 2009.

M. J. Mcconville, R. Wetherbee, and A. Bacic, Subcellular location and composition of the wall and secreted extracellular sulphated polysaccharides/proteoglycans of the diatom Stauroneis amphioxys Gregory, Protoplasma, vol.206, pp.188-200, 1999.

L. K. Medlin, A review of the evolution of the diatoms -a total approach using molecules, morphology and geology, Polish Academy of Sciences, 2000.

N. S. Melo, M. Nimtz, H. S. Conradt, P. S. Fevereiro, C. et al., Identification of the human Lewis(a) carbohydrate motif in a secretory peroxidase from a plant cell suspension culture (Vaccinium myrtillus L, FEBS Lett, vol.415, pp.186-191, 1997.

M. Metzler, A. Gertz, M. Sarkar, H. Schachter, J. W. Schrader et al., Complex asparagine-linked oligosaccharides are required for morphogenic events during postimplantation development, EMBO J, vol.13, pp.2056-2065, 1994.

B. Meusser, C. Hirsch, E. Jarosch, and T. Sommer, ERAD: the long road to destruction, Nat. Cell Biol, vol.7, pp.766-772, 2005.

H. A. Meyer, H. Grau, R. Kraft, S. Kostka, S. Prehn et al., , 2000.

, Mammalian Sec61 is associated with Sec62 and Sec63, J. Biol. Chem, vol.275, pp.14550-14557

M. Miyahara, M. Aoi, N. Inoue-kashino, Y. Kashino, and K. Ifuku, Highly Efficient Transformation of the Diatom Phaeodactylum tricornutum by Multi-Pulse Electroporation, Biosci. Biotechnol. Biochem, 2013.

E. Miyoshi, K. Noda, Y. Yamaguchi, S. Inoue, Y. Ikeda et al., The alpha1-6-fucosyltransferase gene and its biological significance, Biochim. Biophys. Acta, vol.1473, pp.9-20, 1999.

E. Miyoshi, K. Moriwaki, and T. Nakagawa, Biological Function of Fucosylation in Cancer Biology, J. Biochem. (Tokyo), vol.143, pp.725-729, 2007.

S. Moeys, J. Frenkel, C. Lembke, J. T. Gillard, V. Devos et al., A sex-inducing pheromone triggers cell cycle arrest and mate attraction in the diatom Seminavis robusta, Sci. Rep, vol.6, 2016.

E. Mohorko, R. Glockshuber, A. , and M. , Oligosaccharyltransferase: the central enzyme of N-linked protein glycosylation, J. Inherit. Metab. Dis, vol.34, pp.869-878, 2011.

M. Molinari, K. K. Eriksson, V. Calanca, C. Galli, P. Cresswell et al., Contrasting functions of calreticulin and calnexin in glycoprotein folding and ER quality control, Mol. Cell, vol.13, pp.125-135, 2004.

R. Mollicone, S. E. Moore, N. Bovin, M. Garcia-rosasco, J. Candelier et al., Activity, Splice Variants, Conserved Peptide Motifs, and Phylogeny of Two New 1,3-Fucosyltransferase Families (FUT10 and FUT11), J. Biol. Chem, vol.284, pp.4723-4738, 2009.

A. Montsant, K. Jabbari, U. Maheswari, and C. Bowler, Comparative Genomics of the Pennate Diatom Phaeodactylum tricornutum, Plant Physiol, vol.137, pp.500-513, 2005.

I. Moore, M. , and A. , Validating the Location of Fluorescent Protein Fusions in the Endomembrane System, The Plant Cell, vol.21, pp.1632-1636, 2009.

V. J. Moreno, J. E. De-moreno, and R. R. Brenner, Biosynthesis of unsaturated fatty acids in the diatom Phaeodactylum tricornutum, Lipids, vol.14, pp.15-19, 1979.

P. Morsomme, C. Prescianotto-baschong, and H. Riezman, The ER v-SNAREs are required for GPI-anchored protein sorting from other secretory proteins upon exit from the ER, J. Cell Biol, vol.162, pp.403-412, 2003.

A. Moustafa, B. Beszteri, U. G. Maier, C. Bowler, K. Valentin et al., Genomic footprints of a cryptic plastid endosymbiosis in diatoms, Science, vol.324, pp.1724-1726, 2009.

B. W. Murray, V. Wittmann, M. D. Burkart, S. Hung, and C. Wong, Mechanism of Human ?-1,3-Fucosyltransferase V: Glycosidic Cleavage Occurs Prior to Nucleophilic Attack ?, Biochemistry (Mosc.), vol.36, pp.823-831, 1997.

F. Mus, J. Toussaint, K. E. Cooksey, M. W. Fields, R. Gerlach et al., Physiological and molecular analysis of carbon source supplementation and pH 275 Références bibliographiques stress-induced lipid accumulation in the marine diatom Phaeodactylum tricornutum, Appl. Microbiol. Biotechnol, vol.97, pp.3625-3642, 2013.

S. Nagai and I. Imai, Enumeration of bacteria in seawater and sediment from the Seto Inland Sea of Japan that promote sperm formation in Coscinodiscus wailesii (Bacillariophyceae), Phycologia, vol.37, pp.363-368, 1998.

A. I. Nesvizhskii, Proteogenomics: concepts, applications and computational strategies, Nat. Methods, vol.11, pp.1114-1125, 2014.

E. Nguema-ona, M. Vicré-gibouin, M. Gotté, B. Plancot, P. Lerouge et al., Cell wall O-glycoproteins and N-glycoproteins: aspects of biosynthesis and function, Front. Plant Sci, vol.5, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01842174

I. M. Nilsson, V. Heijne, and G. , Determination of the distance between the oligosaccharyltransferase active site and the endoplasmic reticulum membrane, J. Biol. Chem, vol.268, pp.5798-5801, 1993.

Y. Niu, Z. Yang, M. Zhang, C. Zhu, W. Yang et al., Transformation of diatom Phaeodactylum tricornutum by electroporation and establishment of inducible selection marker, BioTechniques, 2012.

C. Noffz, S. Keppler-ross, and N. Dean, Hetero-oligomeric interactions between early glycosyltransferases of the dolichol cycle, Glycobiology, vol.19, pp.472-478, 2009.

T. Nolan, R. E. Hands, and S. A. Bustin, Quantification of mRNA using real-time RT-PCR, Nat. Protoc, vol.1, pp.1559-1582, 2006.

T. M. Norden-krichmar, A. E. Allen, T. Gaasterland, and M. Hildebrand, Characterization of the Small RNA Transcriptome of the Diatom, Thalassiosira pseudonana, PLoS ONE, vol.6, 2011.

M. Nymark, K. C. Valle, T. Brembu, K. Hancke, P. Winge et al., An integrated analysis of molecular acclimation to high light in the marine diatom Phaeodactylum tricornutum, PloS One, vol.4, p.7743, 2009.

S. Olivari, T. Cali, K. E. Salo, P. Paganetti, L. W. Ruddock et al., EDEM1 regulates ER-associated degradation by accelerating de-mannosylation of folding-defective polypeptides and by inhibiting their covalent aggregation, Biochem. Biophys. Res. Commun, vol.349, pp.1278-1284, 2006.

M. K. O'reilly, G. Zhang, and B. Imperiali, In Vitro Evidence for the Dual Function of Alg2 and Alg11: Essential Mannosyltransferases in N-Linked Glycoprotein Biosynthesis ?, Biochemistry (Mosc.), vol.45, pp.9593-9603, 2006.

R. Oriol, R. Mollicone, A. Cailleau, L. Balanzino, and C. Breton, Divergent evolution of fucosyltransferase genes from vertebrates, invertebrates, and bacteria, Glycobiology, vol.9, pp.323-334, 1999.
URL : https://hal.archives-ouvertes.fr/hal-00309806

P. Orlean, C. Albright, and P. W. Robbins, Cloning and sequencing of the yeast gene for dolichol phosphate mannose synthase, an essential protein, J. Biol. Chem, vol.263, pp.17499-17507, 1988.

S. Pagny, F. Bouissonnie, M. Sarkar, M. L. Follet-gueye, A. Driouich et al., Structural requirements for Arabidopsis ?1, 2-xylosyltransferase activity and targeting to the Golgi, Plant J, vol.33, pp.189-203, 2003.

A. S. Palma, V. A. Morais, A. V. Coelho, C. , and J. , Effect of the manganese ion on human ?3/4 fucosyltransferase III activity, Biometals, vol.17, pp.35-43, 2004.

A. R. Paolacci, O. A. Tanzarella, E. Porceddu, and M. Ciaffi, Identification and validation of reference genes for quantitative RT-PCR normalization in wheat, BMC Mol. Biol, vol.10, p.11, 2009.

J. Parkinson, G. , and R. , Beyond micromachining: the potential of diatoms, Trends Biotechnol, vol.17, pp.190-196, 1999.

F. Parlati, J. A. Mcnew, R. Fukuda, R. Miller, T. H. Söllner et al., , 2000.

, Topological restriction of SNARE-dependent membrane fusion, Nature, vol.407, pp.194-198

A. J. Parodi, Role of N-oligosaccharide endoplasmic reticulum processing reactions in glycoprotein folding and degradation, Biochem. J, vol.348, pp.1-13, 2000.

K. Paschinger, D. Rendi?, G. Lochnit, V. Jantsch, and I. B. Wilson, Molecular Basis of Anti-horseradish Peroxidase Staining in Caenorhabditis elegans, J. Biol. Chem, vol.279, pp.49588-49598, 2004.

J. C. Paulson, Glycoproteins: what are the sugar chains for ?, Trends Biochem. Sci, vol.14, pp.272-276, 1989.

A. Petrescu, A. Milac, S. M. Petrescu, R. A. Dwek, and M. R. Wormald, Statistical analysis of the protein environment of N-glycosylation sites: implications for occupancy, structure, and folding, Glycobiology, vol.14, pp.103-114, 2003.

M. W. Pfaffl, A new mathematical model for relative quantification in real-time RT-PCR, Nucleic Acids Res, vol.29, p.45, 2001.

M. W. Pfaffl, A. Tichopad, C. Prgomet, and T. P. Neuvians, Determination of stable housekeeping genes, differentially regulated target genes and sample integrity: BestKeeper--Excel-based tool using pair-wise correlations, Biotechnol. Lett, vol.26, pp.509-515, 2004.

J. D. Pickett-heaps, A. M. Schmid, and D. H. Tippit, Cell division in diatoms: A translation of part of Robert Lauterborn's treatise of 1896 with some modern confirmatory observations, Protoplasma, vol.120, pp.132-154, 1984.

M. Pieren, C. Galli, A. Denzel, and M. Molinari, The Use of Calnexin and Calreticulin by Cellular and Viral Glycoproteins, J. Biol. Chem, vol.280, pp.28265-28271, 2005.

W. H. Poll, . Van-de, E. G. Vrieling, and W. W. Gieskes, Location and expression of frustulins in the pennate diatoms Cylindrotheca fusiformis, Navicula pelliculosa, and Navicula salinarum (Bacillariophyceae), J. Phycol, vol.35, pp.1044-1053, 1999.

J. Prihoda, A. Tanaka, W. B. De-paula, J. F. Allen, L. Tirichine et al., , 2012.

, Chloroplast-mitochondria cross-talk in diatoms, J. Exp. Bot, vol.63, pp.1543-1557

, Références bibliographiques

M. Pucic, A. Knezevic, J. Vidic, B. Adamczyk, M. Novokmet et al., High Throughput Isolation and Glycosylation Analysis of IgG-Variability and Heritability of the IgG Glycome in Three Isolated Human Populations, Mol. Cell. Proteomics, vol.10, 2011.

I. Reichardt, Y. Stierhof, U. Mayer, S. Richter, H. Schwarz et al., Plant Cytokinesis Requires De Novo Secretory Trafficking but Not Endocytosis, Curr. Biol, vol.17, pp.2047-2053, 2007.

G. Reiss, S. Te-heesen, J. Zimmerman, P. Robbins, A. et al., Isolation of the ALG6 locus of Saccharomyces cerevisiae required for glucosylation in the N-linked glycosylation pathway, Glycobiology, vol.6, pp.493-498, 1996.

C. Ritter and A. Helenius, Recognition of local glycoprotein misfolding by the ER folding sensor UDP-glucose: glycoprotein glucosyltransferase, Nat. Struct. Mol. Biol, vol.7, pp.278-280, 2000.

M. D. Robinson and G. K. Smyth, Moderated statistical tests for assessing differences in tag abundance, Bioinforma. Oxf. Engl, vol.23, pp.2881-2887, 2007.

A. Rogato, H. Richard, A. Sarazin, B. Voss, S. C. Navarro et al., The diversity of small non-coding RNAs in the diatom Phaeodactylum tricornutum, BMC Genomics, vol.15, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01064372

F. E. Round, R. M. Crawford, and D. G. Mann, The Diatoms: biology & morphology of the genera, 1990.

J. S. Rush and C. J. Waechter, Transmembrane movement of a water-soluble analogue of mannosylphosphoryldolichol is mediated by an endoplasmic reticulum protein, J. Cell Biol, vol.130, pp.529-536, 1995.

J. S. Rush, K. Van-leyen, O. Ouerfelli, B. Wolucka, and C. J. Waechter, Transbilayer movement of Glc-P-dolichol and its function as a glucosyl donor: protein-mediated transport of a water-soluble analog into sealed ER vesicles from pig brain, Glycobiology, vol.8, pp.1195-1205, 1998.

S. R. Rushforth, J. R. Johansen, and D. L. Sorensen, Occurrence of Phaeodactylum tricornutum in the Great Salt Lake, Gt. Basin Nat, vol.48, pp.324-326, 1988.

C. Saint-jore-dupas, A. Nebenführ, A. Boulaflous, M. Follet-gueye, C. Plasson et al., Plant N-glycan processing enzymes employ different targeting mechanisms for their spatial arrangement along the secretory pathway, Plant Cell Online, vol.18, pp.3182-3200, 2006.
URL : https://hal.archives-ouvertes.fr/hal-01942851

S. Sanyal and A. K. Menon, Stereoselective transbilayer translocation of mannosyl phosphoryl dolichol by an endoplasmic reticulum flippase, Proc. Natl. Acad. Sci, vol.107, pp.11289-11294, 2010.

S. Sato, G. Beakes, M. Idei, T. Nagumo, and D. G. Mann, Novel Sex Cells and Evidence for Sex Pheromones in Diatoms, PLoS ONE, vol.6, 2011.

T. Satoh, K. Suzuki, T. Yamaguchi, and K. Kato, Structural Basis for Disparate SugarBinding Specificities in the Homologous Cargo Receptors ERGIC-53 and VIP36, PLoS ONE, vol.9, p.87963, 2014.

A. Sawaguchi, S. Ide, Y. Goto, J. Kawano, T. Oinuma et al., A simple contrast enhancement by potassium permanganate oxidation for Lowicryl K4M ultrathin sections prepared by high pressure freezing/freeze substitution, J. Microsc, vol.201, pp.77-83, 2001.

C. N. Scanlan, D. R. Burton, and R. A. Dwek, Making autoantibodies safe, Proc. Natl. Acad. Sci. 105, pp.4081-4082, 2008.

B. E. Schaub, E. G. Berger, and J. Rohrer, Dissection of a novel molecular determinant mediating Golgi to trans-Golgi network transition, Cell. Mol. Life Sci, vol.65, pp.3677-3687, 2008.

B. Schiller, A. Hykollari, S. Yan, K. Paschinger, and I. B. Wilson, Complicated Nlinked glycans in simple organisms, Biol. Chem, vol.393, 2012.

J. Schoberer and R. Strasser, Sub-Compartmental Organization of Golgi-Resident NGlycan Processing Enzymes in Plants, Mol. Plant, vol.4, pp.220-228, 2011.

J. Schoberer, E. Liebminger, S. W. Botchway, R. Strasser, and C. Hawes, TimeResolved Fluorescence Imaging Reveals Differential Interactions of N-Glycan Processing Enzymes across the Golgi Stack in Planta, Plant Physiol, vol.161, pp.1737-1754, 2013.

J. D. Schrag, J. J. Bergeron, Y. Li, S. Borisova, M. Hahn et al., The structure of calnexin, an ER chaperone involved in quality control of protein folding, Mol. Cell, vol.8, pp.633-644, 2001.

F. Schwarz, A. , and M. , Mechanisms and principles of N-linked protein glycosylation, Curr. Opin. Struct. Biol, vol.21, pp.576-582, 2011.

K. Shailubhai, B. S. Pukazhenthi, E. S. Saxena, G. M. Varma, and I. K. Vijay, , 1991.

, Glucosidase I, a transmembrane endoplasmic reticular glycoprotein with a luminal catalytic domain, J. Biol. Chem, vol.266, pp.16587-16593

H. J. Sharpe, T. J. Stevens, and S. Munro, A Comprehensive Comparison of Transmembrane Domains Reveals Organelle-Specific Properties, Cell, vol.142, pp.158-169, 2010.

D. Shental-bechor and Y. Levy, Effect of glycosylation on protein folding: a close look at thermodynamic stabilization, Proc. Natl. Acad. Sci. 105, pp.8256-8261, 2008.

T. Shinkawa, K. Nakamura, N. Yamane, E. Shoji-hosaka, Y. Kanda et al., The Absence of Fucose but Not the Presence of Galactose or Bisecting N-Acetylglucosamine of Human IgG1 Complex-type Oligosaccharides Shows the Critical Role of Enhancing Antibody-dependent Cellular Cytotoxicity, J. Biol. Chem, vol.278, pp.3466-3473, 2003.

S. Shrimal, B. G. Ng, M. Losfeld, R. Gilmore, and H. H. Freeze, Mutations in STT3A and STT3B cause two congenital disorders of glycosylation, Hum. Mol. Genet, vol.22, pp.4638-4645, 2013.

M. Siaut, M. Heijde, M. Mangogna, A. Montsant, S. Coesel et al., Molecular toolbox for studying diatom biology in Phaeodactylum tricornutum, Gene, vol.406, pp.23-35, 2007.

S. Singh and A. Mittal, Transmembrane Domain Lengths Serve as Signatures of, Organismal Complexity and Viral Transport Mechanisms. Sci. Rep, vol.6, p.22352, 2016.

M. D. Snider and O. C. Rogers, Transmembrane movement of oligosaccharide-lipids during glycoprotein synthesis, Cell, vol.36, pp.753-761, 1984.

M. Sousa and A. J. Parodi, The molecular basis for the recognition of misfolded glycoproteins by the UDP-Glc: glycoprotein glucosyltransferase, EMBO J, vol.14, p.4196, 1995.

I. Stagljar, S. Te-heesen, A. , and M. , New phenotype of mutations deficient in glucosylation of the lipid-linked oligosaccharide: cloning of the ALG8 locus, Proc. Natl. Acad. Sci, vol.91, pp.5977-5981, 1994.

M. S. Stanley and J. A. Callow, Whole cell adhesion strength of morphotypes and isolates of Phaeodactylum tricornutum (Bacillariophyceae), Eur. J. Phycol, vol.42, pp.191-197, 2007.

E. Staudacher and L. März, Strict order of (Fuc to Asn-linked GlcNAc) fucosyltransferases forming core-difucosylated structures, Glycoconj. J, vol.15, pp.355-360, 1998.

R. Strasser, Plant protein glycosylation, Glycobiology, vol.26, pp.926-939, 2016.

R. Strasser, J. Mucha, H. Schwihla, F. Altmann, J. Glössl et al., Molecular cloning and characterization of cDNA coding for ?1, 2N-acetylglucosaminyltransferase I (GlcNAc-TI) from Nicotiana tabacum, Glycobiology, vol.9, pp.779-785, 1999.

R. Strasser, J. Schoberer, C. Jin, J. Glössl, L. Mach et al., Molecular cloning and characterization of Arabidopsis thaliana Golgi ?-mannosidase II, a key enzyme in the formation of complex N-glycans in plants, Plant J, vol.45, pp.789-803, 2006.

R. Strasser, J. S. Bondili, J. Schoberer, B. Svoboda, E. Liebminger et al., Enzymatic Properties and Subcellular Localization of Arabidopsis beta-N-Acetylhexosaminidases, Plant Physiol, vol.145, pp.5-16, 2007.

D. Studer, W. Graber, A. Al-amoudi, and P. Eggli, A new approach for cryofixation by high-pressure freezing, J. Microsc, vol.203, pp.285-294, 2001.

A. Sturm, K. D. Johnson, T. Szumilo, A. D. Elbein, and M. J. Chrispeels, Subcellular localization of glycosidases and glycosyltransferases involved in the processing of N-linked oligosaccharides, Plant Physiol, vol.85, pp.741-745, 1987.

M. Sumper and N. Kröger, Silica formation in diatoms: the function of long-chain polyamines and silaffins, J. Mater. Chem, vol.14, p.2059, 2004.

H. Sun, S. Lin, T. Ko, J. Pan, C. Liu et al., Structure and Mechanism of Helicobacter pylori Fucosyltransferase: A BASIS FOR LIPOPOLYSACCHARIDE VARIATION AND INHIBITOR DESIGN, J. Biol. Chem, vol.282, pp.9973-9982, 2007.

T. Szumilo, G. P. Kaushal, H. Hori, and A. D. Elbein, Purification and properties of a glycoprotein processing ?-mannosidase from mung bean seedlings, Plant Physiol, vol.81, pp.383-389, 1986.

V. W. Tai, .. Imperiali, and B. , Substrate Specificity of the Glycosyl Donor for Oligosaccharyl Transferase, J. Org. Chem, vol.66, pp.6217-6228, 2001.

E. Takahashi, J. Ledauphin, D. Goux, and F. Orvain, Optimising extraction of extracellular polymeric substances (EPS) from benthic diatoms: comparison of the efficiency of six EPS extraction methods, Mar. Freshw. Res, vol.60, pp.1201-1210, 2009.

R. M. Tashiro, J. G. Philips, and C. S. Winefield, Identification of suitable grapevine reference genes for qRT-PCR derived from heterologous species, Mol. Genet. Genomics MGG, vol.291, pp.483-492, 2016.

D. C. Tessier, D. Dignard, A. Zapun, A. Radominska-pandya, A. J. Parodi et al., Cloning and characterization of mammalian UDP-glucose glycoprotein: glucosyltransferase and the development of a specific substrate for this enzyme, Glycobiology, vol.10, pp.403-412, 2000.

B. Tesson, C. Gaillard, M. , and V. , Insights into the polymorphism of the diatom Phaeodactylum tricornutum Bohlin, 1952.

K. Tezuka, M. Hayashi, H. Ishihara, T. Akazawa, and N. Takahashi, Studies on synthetic pathway of xylose-containing N-linked oligosaccharides deduced from substrate specificities of the processing enzymes in sycamore cells (Acer pseudoplatanus L.), Eur. J. Biochem, vol.203, pp.401-413, 1992.

O. Thellin, B. Elmoualij, E. Heinen, and W. Zorzi, A decade of improvements in quantification of gene expression and internal standard selection, Biotechnol. Adv, vol.27, pp.323-333, 2009.

C. Trapnell, L. Pachter, and S. L. Salzberg, TopHat: discovering splice junctions with RNA-Seq, Bioinforma. Oxf. Engl, vol.25, pp.1105-1111, 2009.

P. Tréguer, D. Nelson, M. Van-bennekom, A. , J. De-master et al., The silica balance in the world ocean: a reestimate, Science, vol.268, pp.375-379, 1995.

E. S. Trombetta, J. F. Simons, and A. Helenius, Endoplasmic reticulum glucosidase II is composed of a catalytic subunit, conserved from yeast to mammals, and a tightly bound noncatalytic HDEL-containing subunit, J. Biol. Chem, vol.271, pp.27509-27516, 1996.

J. Valenzuela, A. Mazurie, R. P. Carlson, R. Gerlach, K. E. Cooksey et al., Potential role of multiple carbon fixation pathways during lipid accumulation in Phaeodactylum tricornutum, Biotechnol. Biofuels, vol.5, p.40, 2012.

J. Vandesompele, K. De-preter, F. Pattyn, B. Poppe, N. Van-roy et al., Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes, Genome Biol, vol.3, pp.1-12, 2002.

, Références bibliographiques

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.10, p.139282, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01841078

A. Varki, Biological roles of oligosaccharides: all of the theories are correct, Glycobiology, vol.3, pp.97-130, 1993.

A. Velasco, L. Hendricks, K. W. Moremen, D. R. Tulsiani, O. Touster et al., Cell type-dependent variations in the subcellular distribution of alpha-mannosidase I and II, J. Cell Biol, vol.122, pp.39-51, 1993.

G. J. Vella, H. Paulsen, and H. Schachter, Control of glycoprotein synthesis. IX. A terminal Man alpha l-3Man beta 1-sequence in the substrate is the minimum requirement for UDP-N-acetyl-D-glucosamine: alpha-D-mannoside (GlcNAc to Man alpha 1-3) beta 2-Nacetylglucosaminyltransferase I, Can. J. Biochem. Cell Biol. Rev. Can. Biochim. Biol. Cell, vol.62, pp.409-417, 1984.

S. S. Vembar and J. L. Brodsky, One step at a time: endoplasmic reticulum-associated degradation, Nat. Rev. Mol. Cell Biol, vol.9, pp.944-957, 2008.

T. Vermassen, C. Van-praet, N. Lumen, K. Decaestecker, D. Vanderschaeghe et al., Urinary prostate protein glycosylation profiling as a diagnostic biomarker for prostate cancer: Prostate Protein Glycosylation in Prostate Cancer, The Prostate, vol.75, pp.314-322, 2015.

B. E. Volcani, Cell wall formation in diatoms: morphogenesis and biochemistry, Silicon and Siliceous Structures in Biological Systems, pp.157-200, 1981.

J. A. Voynow, R. S. Kaiser, T. F. Scanlin, and M. C. Glick, Purification and characterization of GDP-L-fucose-N-acetyl beta-D-glucosaminide alpha 1----6fucosyltransferase from cultured human skin fibroblasts. Requirement of a specific biantennary oligosaccharide as substrate, J. Biol. Chem, vol.266, pp.21572-21577, 1991.

E. G. Vrieling, W. W. Gieskes, and T. P. Beelen, Silicon deposition in diatoms: control by the pH inside the silicon deposition vesicle, J. Phycol, vol.35, pp.548-559, 1999.

T. De-vries, R. M. Knegtel, E. H. Holmes, and B. A. Macher, Fucosyltransferases: structure/function studies, Glycobiology, vol.11, pp.119-128, 2001.

I. Wada, S. Imai, M. Kai, F. Sakane, and H. Kanoh, Chaperone Function of Calreticulin When Expressed in the Endoplasmic Reticulum as the Membrane-anchored and Soluble Forms, J. Biol. Chem, vol.270, pp.20298-20304, 1995.

X. Wang, Core Fucosylation Regulates Epidermal Growth Factor Receptor-mediated Intracellular Signaling, J. Biol. Chem, vol.281, pp.2572-2577, 2005.

X. Wang, T. Weldeghiorghis, G. Zhang, B. Imperiali, and J. H. Prestegard, Solution Structure of Alg13: The Sugar Donor Subunit of a Yeast N-Acetylglucosamine Transferase, Structure, vol.16, pp.965-975, 2008.

Y. Wang, T. Fukuda, T. Isaji, J. Lu, W. Gu et al., Loss of ?1,6-fucosyltransferase suppressed liver regeneration: implication of core fucose in the regulation of growth factor receptor-mediated cellular signaling, Sci. Rep, vol.5, p.8264, 2015.

S. Watanabe, A. Kakudo, M. Ohta, K. Mita, K. Fujiyama et al., Molecular cloning and characterization of the ?-glucosidase II from Bombyx mori and Spodoptera frugiperda, Insect Biochem. Mol. Biol, vol.43, pp.319-327, 2013.

F. Wendler and S. Tooze, Syntaxin 6: the promiscuous behaviour of a SNARE protein, Traffic Cph. Den, vol.2, pp.606-611, 2001.

R. Wetherbee, BIOMINERALIZATION: The Diatom Glasshouse, Science, vol.298, pp.547-547, 2002.

P. Whitley, I. Nilsson, and G. Heijne, A Nascent Secretory Protein 5 Traverse the Ribosome/Endoplasmic Reticulum Translocase Complex as an Extended Chain, J. Biol. Chem, vol.271, pp.6241-6244, 1996.

S. J. Wijeyesakere, J. K. Gagnon, K. Arora, C. L. Brooks, and M. Raghavan, Regulation of calreticulin-major histocompatibility complex (MHC) class I interactions by ATP, Proc. Natl. Acad. Sci, vol.112, pp.5608-5617, 2015.

S. Wildt and T. U. Gerngross, The humanization of N-glycosylation pathways in yeast, Nat. Rev. Microbiol, vol.3, pp.119-128, 2005.

A. Willis, A. Chiovitti, T. M. Dugdale, and R. Wetherbee, Characterization of the extracellular matrix of Phaeodactylum tricornutum (Bacillariophyceae): structure, composition, and adhesive characteristics, J. Phycol, pp.937-949, 2013.

D. Wilson, The triradiate and other forms of Nitzschia closterum, 1946.

I. B. Wilson, Glycosylation of proteins in plants and invertebrates, Curr. Opin. Struct. Biol, vol.12, pp.569-577, 2002.

I. B. Wilson, The class I ?1,2-mannosidases of Caenorhabditis elegans, Glycoconj. J, vol.29, pp.173-179, 2012.

I. B. Wilson, J. E. Harthill, N. P. Mullin, D. A. Ashford, and F. Altmann, Core ?1, 3-fucose is a key part of the epitope recognized by antibodies reacting against plant N-linked oligosaccharides and is present in a wide variety of plant extracts, Glycobiology, vol.8, pp.651-661, 1998.

I. B. Wilson, D. Rendi?, A. Freilinger, J. Dumi?, F. Altmann et al., Cloning and expression of cDNAs encoding ?1, 3-fucosyltransferase homologues from Arabidopsis thaliana, Biochim. Biophys. Acta BBA-Gen. Subj, vol.1527, pp.88-96, 2001.

J. R. Wilson, D. Williams, and H. Schachter, The control of glycoprotein synthesis: Nacetylglucosamine linkage to a mannose residue as a signal for the attachment of L-fucose to the asparagine-linked N-acetylglucosamine residue of glycopeptide from alpha1-acid glycoprotein, Biochem. Biophys. Res. Commun, vol.72, pp.909-916, 1976.

D. Winckelmann, F. Bleeke, B. Thomas, C. Elle, and G. Klöck, Open pond cultures of indigenous algae grown on non-arable land in an arid desert using wastewater, Int. Aquat. Res, vol.7, pp.221-233, 2015.

A. Z. Worden, M. J. Follows, S. J. Giovannoni, S. Wilken, A. E. Zimmerman et al., Rethinking the marine carbon cycle: Factoring in the multifarious lifestyles of microbes, Science, vol.347, pp.1257594-1257594, 2015.

X. Xiao, X. Wu, J. Ma, P. Li, T. Li et al., Systematic assessment of reference genes for RT-qPCR across plant species under salt stress and drought stress, Acta Physiol. Plant, vol.37, 2015.

F. Xie, P. Xiao, D. Chen, L. Xu, and B. Zhang, miRDeepFinder: a miRNA analysis tool for deep sequencing of plant small RNAs, Plant Mol. Biol, 2012.

Q. Yan and W. J. Lennarz, Oligosaccharyltransferase: A Complex Multisubunit Enzyme of the Endoplasmic Reticulum, Biochem. Biophys. Res. Commun, vol.266, pp.684-689, 1999.

Q. Yan and W. J. Lennarz, Studies on the Function of Oligosaccharyl Transferase Subunits: Stt3p IS DIRECTLY INVOLVED IN THE GLYCOSYLATION PROCESS, J. Biol. Chem, vol.277, pp.47692-47700, 2002.

Z. Yang, Y. Niu, Y. Ma, J. Xue, M. Zhang et al., Molecular and cellular mechanisms of neutral lipid accumulation in diatom following nitrogen deprivation, Biotechnol. Biofuels, vol.6, p.67, 2013.

A. Yool and T. Tyrrell, Role of diatoms in regulating the ocean's silicon cycle, Glob. Biogeochem. Cycles, vol.17, 2003.

C. Zhang and H. Hu, High-efficiency nuclear transformation of the diatom Phaeodactylum tricornutum by electroporation, Mar. Genomics, 2013.

H. Zhang, M. Shahbazi, E. M. Mäkilä, T. H. Da-silva, R. L. Reis et al., Diatom silica microparticles for sustained release and permeation enhancement following oral delivery of prednisone and mesalamine, Biomaterials, vol.34, pp.9210-9219, 2013.

P. Zhao, W. Gu, S. Wu, A. Huang, L. He et al., Silicon enhances the growth of Phaeodactylum tricornutum Bohlin under green light and low temperature, 2014.

X. Zhao, H. Yang, W. Liu, X. Duan, W. Shang et al., Sec22 regulates endoplasmic reticulum morphology but not autophagy and is required for eye development in Drosophila, J. Biol. Chem, vol.290, pp.7943-7951, 2015.

D. F. Zielinska, F. Gnad, J. R. Wi?niewski, and M. Mann, Precision Mapping of an In Vivo N-Glycoproteome Reveals Rigid Topological and Sequence Constraints, Cell, vol.141, pp.897-907, 2010.

D. F. Zielinska, F. Gnad, K. Schropp, J. R. Wi?niewski, and M. Mann, Mapping NGlycosylation Sites across Seven Evolutionarily Distant Species Reveals a Divergent Substrate Proteome Despite a Common Core Machinery, Mol. Cell, vol.46, pp.542-548, 2012.

C. Zurzolo and C. Bowler, Exploring Bioinorganic Pattern Formation in Diatoms. A Story of Polarized Trafficking, PLANT Physiol, vol.127, pp.1339-1345, 2001.

. R-É-s-u-m-É-/-a-b-s-t-r-a-c-t,

L. , Dans ces travaux, nous avons poursuivi l'exploration de la voie de N-glycosylation de la diatomée modèle P. tricornutum en focalisant spécifiquement notre travail sur la caractérisation de trois ?1,3-fucosyltransférases putatives. Nos analyses ont permis de remettre en cause l'annotation pour deux des gènes codant pour ces fucosyltransférases et d'en proposer de nouvelles. Afin de réaliser l'étude de la localisation subcellulaire de ces glycosyltransférases, nous avons utilisé une stratégie de sur-expression de ces protéines fusionnées avec une étiquette. Les observations menées par des approches de microscopie confocale et de microscopie électronique à transmission ont permises de mettre en évidence la localisation subcellulaire golgienne de ces enzymes, ainsi que de trois autres glycosyltransférases utilisées comme témoin dans ce travail, N-glycosylation est un événement co-et post-traductionnel majeur de la synthèse protéique chez les eucaryotes. Actuellement, peu d'informations concernant ce processus sont disponibles chez les microalgues

, Afin d'apporter de nouvelles réponses, nous avons réalisé une analyse comparative du transcriptome des trois morphotypes par une approche transcriptomique à haut débit appelée RNASeq pour RNA-Sequencing. Enfin, en plus d'élargir les connaissances sur la morphogénèse de P. tricornutum, les données générées ont également permis d, P. tricornutum est une diatomée pléïomorphique qui possède trois morphotypes majeurs différents appelés ovale, fusiforme et triradié

. Actually, In this work, we continued the exploration of the N-glycosylation pathway in the model diatom P. tricornutum by focusing especially on the characterization of three putative ?1,3-fucosyltransferases. Our analyzes question the annotation for two of the genes encoding for these fucosyltransferases and propose new ones. In order to achieve the subcellular localization of these glycosyltransferases, we used an over-expressed fusion tag proteins strategy combined together with microscopy approaches. Observations performed by confocal microscopy and TEM allowed us to demonstrate the Golgi subcellular localization of these enzymes

, The transition from one morphotype to another strongly depends on the environmental conditions. However, little information is available regarding the mechanisms and physiological significance of this morphogenesis. In order to provide new knowledge related to the morphogenesis in P. tricornutm, we conducted a comparative transcriptome analysis of the three morphotypes by an RNA-Seq approach. Besides expanding our knowledge about P. tricornutum morphogenesis, P. tricornutum possess three different morphotypes named oval, fusiform and triradiate

, Mots-clés : N-glycosylation, Phaeodactylum tricornutum, appareil de Golgi, glycosyltransferase, RNA-Seq, diatomées, transcriptome, localisation subcellulaire, morphogenèse