J. D. Watson and F. H. Crick, Molecular structure of nucleic acids. A structure for deoxyribose nucleic acid, Rev Investig Clin Organo Hosp Enfermedades Nutr. avr, vol.55, issue.2, pp.108-117, 1953.

R. Dahm, Discovering DNA: Friedrich Miescher and the early years of nucleic acid research, Hum Genet. janv, vol.122, issue.6, pp.565-81, 2008.

S. Quinodoz and M. Guttman, Long non-coding RNAs: An emerging link between gene regulation and nuclear organization, Trends Cell Biol. nov, vol.24, issue.11, pp.651-63, 2014.

M. Rutenberg-schoenberg, A. N. Sexton, and M. D. Simon, The Properties of Long Noncoding RNAs that Regulate Chromatin, Annu Rev Genomics Hum Genet. 21 avr, 2016.

L. D. Sacco, A. Baldassarre, and A. Masotti, Bioinformatics Tools and Novel Challenges in Long Non-Coding RNAs (lncRNAs) Functional Analysis, Int J Mol Sci. 23 déc, vol.13, issue.1, pp.97-114, 2011.

C. A. Hutchison, DNA sequencing: bench to bedside and beyond, Nucleic Acids Res. 1 sept, vol.35, issue.18, pp.6227-6264, 2007.

R. W. Holley, J. Apgar, G. A. Everett, J. T. Madison, M. Marquisee et al., STRUCTURE OF A RIBONUCLEIC ACID. Science. 19 mars, vol.147, issue.3664, pp.1462-1467, 1965.

F. Sanger, G. G. Brownlee, and B. G. Barrell, A two-dimensional fractionation procedure for radioactive nucleotides, J Mol Biol. sept, vol.13, issue.2, pp.373-98, 1965.

G. G. Brownlee and F. Sanger, Nucleotide sequences from the low molecular weight ribosomal RNA of Escherichia coli, J Mol Biol. 14 févr, vol.23, issue.3, pp.337-53, 1967.

S. Cory, K. A. Marcker, S. K. Dube, and B. F. Clark, Primary structure of a methionine transfer RNA from Escherichia coli, Nature. 7 déc, vol.220, issue.5171, pp.1039-1079, 1968.

S. K. Dube, K. A. Marcker, B. F. Clark, and C. S. , Nucleotide sequence of N-formyl-methionyltransfer RNA, Nature. 20 avr, vol.218, issue.5138, pp.232-235, 1968.

H. M. Goodman, J. Abelson, A. Landy, S. Brenner, and J. D. Smith, Amber suppression: a nucleotide change in the anticodon of a tyrosine transfer RNA, Nature. 16 mars, vol.217, issue.5133, pp.1019-1043, 1968.

J. M. Adams, P. G. Jeppesen, F. Sanger, and B. G. Barrell, Nucleotide sequence from the coat protein cistron of R17 bacteriophage RNA, Nature. 6 sept, vol.223, issue.5210, pp.1009-1023, 1969.

M. Jou, W. Haegeman, G. Ysebaert, M. Fiers, and W. , Nucleotide sequence of the gene coding for the bacteriophage MS2 coat protein, Nature. 12 mai, vol.237, issue.5350, pp.82-90, 1972.

W. Fiers, R. Contreras, F. Duerinck, G. Haegeman, D. Iserentant et al., Complete nucleotide sequence of bacteriophage MS2 RNA: primary and secondary structure of the replicase gene, Nature. 8 avr, vol.260, issue.5551, pp.500-507, 1976.

R. Wu, Nucleotide sequence analysis of DNA, Nature New Biol. 19 avr, vol.236, issue.68, pp.198-200, 1972.

R. Wu and A. D. Kaiser, Structure and base sequence in the cohesive ends of bacteriophage lambda DNA, J Mol Biol. 14 août, vol.35, issue.3, pp.523-560, 1968.

F. Sanger and A. R. Coulson, A rapid method for determining sequences in DNA by primed synthesis with DNA polymerase, J Mol Biol. 25 mai, vol.94, issue.3, pp.441-449, 1975.

A. M. Maxam and W. Gilbert, A new method for sequencing DNA, Proc Natl Acad Sci U S A. févr, vol.74, issue.2, pp.560-564, 1977.

F. Sanger, S. Nicklen, and A. R. Coulson, DNA sequencing with chain-terminating inhibitors, Proc Natl Acad Sci U S A. déc, vol.74, issue.12, pp.5463-5470, 1977.

L. M. Smith, S. Fung, M. W. Hunkapiller, T. J. Hunkapiller, and L. E. Hood, The synthesis of oligonucleotides containing an aliphatic amino group at the 5' terminus: synthesis of fluorescent DNA primers for use in DNA sequence analysis, Nucleic Acids Res. 11 avr, vol.13, issue.7, pp.2399-412, 1985.

W. Ansorge, B. S. Sproat, J. Stegemann, and C. Schwager, A non-radioactive automated method for DNA sequence determination, J Biochem Biophys Methods. déc, vol.13, issue.6, pp.315-338, 1986.

W. Ansorge, B. Sproat, J. Stegemann, C. Schwager, and M. Zenke, Automated DNA sequencing: ultrasensitive detection of fluorescent bands during electrophoresis, Nucleic Acids Res. 11 juin, vol.15, issue.11, pp.4593-602, 1987.

J. M. Prober, G. L. Trainor, R. J. Dam, F. W. Hobbs, C. W. Robertson et al., A system for rapid DNA sequencing with fluorescent chain-terminating dideoxynucleotides, Science, vol.238, issue.4825, pp.336-377, 1987.

H. Kambara, T. Nishikawa, Y. Katayama, and T. Yamaguchi, Optimization of Parameters in a DNA Sequenator Using Fluorescence Detection, Bio/Technology. juill, vol.6, issue.7, p.816, 1988.

H. Swerdlow and R. Gesteland, Capillary gel electrophoresis for rapid, high resolution DNA sequencing, Nucleic Acids Res. 25 mars, vol.18, issue.6, pp.1415-1424, 1990.

T. Hunkapiller, R. J. Kaiser, B. F. Koop, and L. Hood, Large-scale and automated DNA sequence determination, Science, vol.254, issue.5028, pp.59-67, 1991.

E. R. Mardis, Next-generation sequencing platforms, Annu Rev Anal Chem Palo Alto Calif, vol.6, pp.287-303, 2013.

D. Dressman, H. Yan, G. Traverso, K. W. Kinzler, and B. Vogelstein, Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations, Proc Natl Acad Sci, vol.100, issue.15, pp.8817-8839, 2003.

J. B. Kim, G. J. Porreca, L. Song, S. C. Greenway, J. M. Gorham et al., Polony multiplex analysis of gene expression (PMAGE) in mouse hypertrophic cardiomyopathy, Science. 8 juin, vol.316, issue.5830, pp.1481-1485, 2007.

J. H. Leamon, W. L. Lee, K. R. Tartaro, J. R. Lanza, G. J. Sarkis et al., A massively parallel PicoTiterPlate based platform for discrete picoliter-scale polymerase chain reactions, Electrophoresis. nov, vol.24, issue.21, pp.3769-77, 2003.

M. Fedurco, A. Romieu, S. Williams, I. Lawrence, and G. Turcatti, BTA, a novel reagent for DNA attachment on glass and efficient generation of solid-phase amplified DNA colonies, Nucleic Acids Res. 9 févr, vol.34, issue.3, p.22, 2006.

T. D. Harris, P. R. Buzby, H. Babcock, E. Beer, J. Bowers et al., Single-molecule DNA sequencing of a viral genome, Science. 4 avr, vol.320, issue.5872, pp.106-115, 2008.

K. V. Voelkerding, S. A. Dames, and J. D. Durtschi, Next-generation sequencing: from basic research to diagnostics, Clin Chem. avr, vol.55, issue.4, pp.641-58, 2009.

D. R. Bentley, S. Balasubramanian, H. P. Swerdlow, G. P. Smith, J. Milton et al., Accurate whole human genome sequencing using reversible terminator chemistry, Nature, vol.456, issue.7218, pp.53-62, 2008.

R. Drmanac, A. B. Sparks, M. J. Callow, A. L. Halpern, N. L. Burns et al., Human genome sequencing using unchained base reads on self-assembling DNA nanoarrays, Science. 1 janv, vol.327, issue.5961, pp.78-81, 2010.

A. E. Tomkinson, S. Vijayakumar, J. M. Pascal, and T. Ellenberger, DNA ligases: structure, reaction mechanism, and function, Chem Rev. févr, vol.106, issue.2, pp.687-99, 2006.

S. Goodwin, J. D. Mcpherson, and W. R. Mccombie, Coming of age: ten years of next-generation sequencing technologies, Nat Rev Genet, vol.17, issue.6, pp.333-51, 2016.

M. Margulies, M. Egholm, W. E. Altman, S. Attiya, J. S. Bader et al., Genome sequencing in microfabricated high-density picolitre reactors, Nature. 15 sept, vol.437, issue.7057, pp.376-80, 2005.

P. Nyrén and A. Lundin, Enzymatic method for continuous monitoring of inorganic pyrophosphate synthesis, Anal Biochem. déc, vol.151, issue.2, pp.504-513, 1985.

J. M. Rothberg, W. Hinz, T. M. Rearick, J. Schultz, W. Mileski et al., An integrated semiconductor device enabling non-optical genome sequencing, Nature. 20 juill, vol.475, issue.7356, pp.348-52, 2011.

J. Guo, N. Xu, Z. Li, S. Zhang, J. Wu et al., Four-color DNA sequencing with 3'-O-modified nucleotide reversible terminators and chemically cleavable fluorescent dideoxynucleotides, Proc Natl Acad Sci, vol.105, issue.27, pp.9145-50, 2008.

J. Ju, D. H. Kim, L. Bi, Q. Meng, X. Bai et al., Four-color DNA sequencing by synthesis using cleavable fluorescent nucleotide reversible terminators, Proc Natl Acad Sci, vol.103, issue.52, pp.19635-19675, 2006.

O. Harismendy, P. C. Ng, R. L. Strausberg, X. Wang, T. B. Stockwell et al., Evaluation of next generation sequencing platforms for population targeted sequencing studies, Genome Biol, vol.10, issue.3, p.32, 2009.

Y. Han, S. Gao, K. Muegge, W. Zhang, and B. Zhou, Advanced Applications of RNA Sequencing and Challenges, Bioinforma Biol Insights, vol.9, issue.1, pp.29-46, 2015.

L. Camarena, V. Bruno, G. Euskirchen, S. Poggio, and M. Snyder, Molecular mechanisms of ethanol-induced pathogenesis revealed by RNA-sequencing, PLoS Pathog. 1 avr, vol.6, issue.4, p.1000834, 2010.

M. Griffith, O. L. Griffith, J. Mwenifumbo, R. Goya, A. S. Morrissy et al., Alternative expression analysis by RNA sequencing, Nat Methods, vol.7, issue.10, pp.843-850, 2010.

E. Picardi, D. S. Horner, M. Chiara, R. Schiavon, G. Valle et al., Large-scale detection and analysis of RNA editing in grape mtDNA by RNA deep-sequencing, Nucleic Acids Res. août, vol.38, issue.14, pp.4755-67, 2010.

B. T. Wilhelm, M. Briau, P. Austin, A. Faubert, G. Boucher et al., RNA-seq analysis of 2 closely related leukemia clones that differ in their self-renewal capacity, Blood. 13 janv, vol.117, issue.2, pp.27-38, 2011.

E. T. Wang, R. Sandberg, S. Luo, I. Khrebtukova, L. Zhang et al., Alternative isoform regulation in human tissue transcriptomes, Nature, vol.456, issue.7221, pp.470-476, 2008.

C. A. Maher, C. Kumar-sinha, X. Cao, S. Kalyana-sundaram, B. Han et al., Transcriptome sequencing to detect gene fusions in cancer, Nature. 5 mars, vol.458, issue.7234, pp.97-101, 2009.

M. F. Berger, J. Z. Levin, K. Vijayendran, A. Sivachenko, X. Adiconis et al., Integrative analysis of the melanoma transcriptome, Genome Res. avr, vol.20, issue.4, pp.413-440, 2010.

J. Supper, C. Gugenmus, J. Wollnik, T. Drueke, M. Scherf et al., Detecting and visualizing gene fusions, Methods San Diego Calif. janv, vol.59, issue.1, pp.24-28, 2013.

L. Conde, P. M. Bracci, R. Richardson, S. B. Montgomery, and C. F. Skibola, Integrating GWAS and expression data for functional characterization of disease-associated SNPs: an application to follicular lymphoma, Am J Hum Genet. 10 janv, vol.92, issue.1, pp.126-156, 2013.

F. Lee and J. Ule, Advances in CLIP Technologies for Studies of Protein-RNA Interactions, Mol Cell. 1 févr, vol.69, issue.3, pp.354-69, 2018.

A. Turchinovich, L. Weiz, A. Langheinz, and B. Burwinkel, Characterization of extracellular circulating microRNA, Nucleic Acids Res. 1 sept, vol.39, issue.16, pp.7223-7256, 2011.

K. C. Vickers, B. T. Palmisano, B. M. Shoucri, R. D. Shamburek, and A. T. Remaley, MicroRNAs are Transported in Plasma and Delivered to Recipient Cells by High-Density Lipoproteins, Nat Cell Biol. avr, vol.13, issue.4, pp.423-456, 2011.

H. Zhang, D. Freitas, H. S. Kim, K. Fabijanic, Z. Li et al., Identification of distinct nanoparticles and subsets of extracellular vesicles by asymmetric flow field-flow fractionation, Nat Cell Biol. mars, vol.20, issue.3, pp.332-375, 2018.

B. György, T. G. Szabó, M. Pásztói, Z. Pál, P. Misják et al., Membrane vesicles, current state-of-the-art: emerging role of extracellular vesicles, Cell Mol Life Sci. août, vol.68, issue.16, pp.2667-88, 2011.

F. Momen-heravi, S. J. Getting, and S. A. Moschos, Extracellular vesicles and their nucleic acids for biomarker discovery, Pharmacol Ther. déc, vol.192, pp.170-87, 2018.

D. W. Greening and R. J. Simpson, Understanding extracellular vesicle diversity -current status, Expert Rev Proteomics. nov, vol.15, issue.11, pp.887-910, 2018.

M. P. Zaborowski, L. Balaj, X. O. Breakefield, and C. P. Lai, Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study, Bioscience. 1 août, vol.65, issue.8, pp.783-97, 2015.

C. Théry, K. W. Witwer, E. Aikawa, M. J. Alcaraz, J. D. Anderson et al., Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines, J Extracell Vesicles, vol.7, issue.1, p.1535750, 2018.

G. Dostert, B. Mesure, P. Menu, and É. Velot, How Do Mesenchymal Stem Cells Influence or Are Influenced by Microenvironment through Extracellular Vesicles Communication? Front, Cell Dev Biol, vol.5, p.6, 2017.

R. Crescitelli, C. Lässer, T. G. Szabó, A. Kittel, M. Eldh et al., Distinct RNA profiles in subpopulations of extracellular vesicles: apoptotic bodies, microvesicles and exosomes, J Extracell Vesicles, vol.2, 2013.

A. Saraste and K. Pulkki, Morphologic and biochemical hallmarks of apoptosis, Cardiovasc Res. févr, vol.45, issue.3, pp.528-565, 2000.

M. Hristov, W. Erl, S. Linder, and P. C. Weber, Apoptotic bodies from endothelial cells enhance the number and initiate the differentiation of human endothelial progenitor cells in vitro, Blood, vol.104, issue.9, pp.2761-2767, 2004.

K. Al-nedawi, B. Meehan, and J. Rak, Microvesicles: messengers and mediators of tumor progression, Cell Cycle Georget Tex. 1 juill, vol.8, issue.13, pp.2014-2022, 2009.

P. A. Holme, N. O. Solum, F. Brosstad, M. Røger, and M. Abdelnoor, Demonstration of plateletderived microvesicles in blood from patients with activated coagulation and fibrinolysis using a filtration technique and western blotting, Thromb Haemost, vol.72, issue.5, pp.666-71, 1994.

G. Van-niel, D. Angelo, G. Raposo, and G. , Shedding light on the cell biology of extracellular vesicles, Nat Rev Mol Cell Biol. avr, vol.19, issue.4, pp.213-241, 2018.

M. Colombo, G. Raposo, and C. Théry, Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles, Annu Rev Cell Dev Biol, vol.30, pp.255-89, 2014.

L. Blanc and M. Vidal, New insights into the function of Rab GTPases in the context of exosomal secretion, Small GTPases, vol.9, issue.1-2, pp.95-106, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02307940

E. Cocucci, G. Racchetti, and J. Meldolesi, Shedding microvesicles: artefacts no more, Trends Cell Biol. févr, vol.19, issue.2, pp.43-51, 2009.

E. Turola, R. Furlan, F. Bianco, M. Matteoli, and C. Verderio, Microglial microvesicle secretion and intercellular signaling, Front Physiol, vol.3, p.149, 2012.

C. D'souza-schorey and J. W. Clancy, Tumor-derived microvesicles: shedding light on novel microenvironment modulators and prospective cancer biomarkers, Genes Dev. 15 juin, vol.26, issue.12, pp.1287-99, 2012.

H. T. Mcmahon and E. Boucrot, Membrane curvature at a glance, J Cell Sci. 15 mars, vol.128, issue.6, pp.1065-70, 2015.

M. M. Kozlov, F. Campelo, N. Liska, L. V. Chernomordik, S. J. Marrink et al., Mechanisms shaping cell membranes, Curr Opin Cell Biol. août, vol.29, pp.53-60, 2014.

G. Raposo and W. Stoorvogel, Extracellular vesicles: exosomes, microvesicles, and friends, J Cell Biol. 18 févr, vol.200, issue.4, pp.373-83, 2013.

N. P. Hessvik and A. Llorente, Current knowledge on exosome biogenesis and release, Cell Mol Life Sci CMLS, vol.75, issue.2, pp.193-208, 2018.

A. V. Savelyeva, E. V. Kuligina, D. N. Bariakin, V. V. Kozlov, E. I. Ryabchikova et al., Variety of RNAs in Peripheral Blood Cells, Plasma, and Plasma Fractions, 2017.

R. C. Rennert, F. H. Hochberg, and B. S. Carter, ExRNA in Biofluids as Biomarkers for Brain Tumors, Cell Mol Neurobiol. avr, vol.36, issue.3, pp.353-60, 2016.

V. Muralidharan-chari, J. W. Clancy, A. Sedgwick, D. Souza-schorey, and C. , Microvesicles: mediators of extracellular communication during cancer progression, J Cell Sci. 15 mai, vol.123, pp.1603-1614, 2010.

D. Toro, J. Herschlik, L. Waldner, C. Mongini, and C. , Emerging roles of exosomes in normal and pathological conditions: new insights for diagnosis and therapeutic applications, Front Immunol, vol.6, p.203, 2015.

C. C. Chen, L. Liu, F. Ma, C. W. Wong, X. E. Guo et al., Elucidation of Exosome Migration across the Blood-Brain Barrier Model In Vitro, Cell Mol Bioeng. déc, vol.9, issue.4, pp.509-538, 2016.

J. Matsumoto, T. Stewart, W. A. Banks, and J. Zhang, The Transport Mechanism of Extracellular Vesicles at the Blood-Brain Barrier, Curr Pharm Des, vol.23, issue.40, pp.6206-6220, 2017.

H. C. Anderson, Vesicles associated with calcification in the matrix of epiphyseal cartilage, J Cell Biol. avr, vol.41, issue.1, pp.59-72, 1969.

B. T. Pan and R. M. Johnstone, Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor, Cell. juill, vol.33, issue.3, pp.967-78, 1983.

J. C. Akers, D. Gonda, R. Kim, B. S. Carter, and C. C. Chen, Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies, J Neurooncol. mai, vol.113, issue.1, pp.1-11, 2013.

A. Beach, H. Zhang, M. Z. Ratajczak, and S. S. Kakar, Exosomes: an overview of biogenesis, composition and role in ovarian cancer, J Ovarian Res. 25 janv, vol.7, p.14, 2014.

W. Möbius, Y. Ohno-iwashita, E. G. Van-donselaar, V. Oorschot, Y. Shimada et al., Immunoelectron microscopic localization of cholesterol using biotinylated and non-cytolytic perfringolysin O, J Histochem Cytochem Off J Histochem Soc. janv, vol.50, issue.1, pp.43-55, 2002.

I. J. White, L. M. Bailey, M. R. Aghakhani, S. E. Moss, and C. E. Futter, EGF stimulates annexin 1-dependent inward vesiculation in a multivesicular endosome subpopulation, EMBO J. 11 janv, vol.25, issue.1, pp.1-12, 2006.

R. Wubbolts, R. S. Leckie, P. Veenhuizen, G. Schwarzmann, W. Möbius et al., Proteomic and biochemical analyses of human B cell-derived exosomes. Potential implications for their function and multivesicular body formation, J Biol Chem. 28 mars, vol.278, issue.13, pp.10963-72, 2003.

K. Trajkovic, C. Hsu, S. Chiantia, L. Rajendran, D. Wenzel et al., Ceramide triggers budding of exosome vesicles into multivesicular endosomes, Science. 29 févr, vol.319, issue.5867, pp.1244-1251, 2008.

J. F. Brouwers, M. Aalberts, J. Jansen, G. Van-niel, M. H. Wauben et al., Distinct lipid compositions of two types of human prostasomes, Proteomics. mai, vol.13, pp.1660-1666, 2013.

A. S. Balkom-bwm-van,-eisele, D. M. Pegtel, S. Bervoets, and M. C. Verhaar, Quantitative and qualitative analysis of small RNAs in human endothelial cells and exosomes provides insights into localized RNA processing, degradation and sorting, J Extracell Vesicles, vol.29, issue.2015

E. Willms, H. J. Johansson, I. Mäger, Y. Lee, K. Blomberg et al., Cells release subpopulations of exosomes with distinct molecular and biological properties, Sci Rep. 2 mars, vol.6, p.22519, 2016.

Z. Andreu and M. Yáñez-mó, Tetraspanins in extracellular vesicle formation and function, Front Immunol, vol.5, p.442, 2014.

C. Gutiérrez-vázquez, C. Villarroya-beltri, M. Mittelbrunn, and F. Sánchez-madrid, Transfer of extracellular vesicles during immune cell-cell interactions, Immunol Rev. janv, vol.251, issue.1, pp.125-167, 2013.

C. Villarroya-beltri, F. Baixauli, C. Gutiérrez-vázquez, F. Sánchez-madrid, M. Mittelbrunn et al., REGULATION OF EXOSOME LOADING. Semin Cancer Biol. oct, vol.28, pp.3-13, 2014.

E. J. Van-der-vlist, G. Arkesteijn, C. Van-de-lest, W. Stoorvogel, E. Nolte-'t-hoen et al., CD4(+) T cell activation promotes the differential release of distinct populations of nanosized vesicles, J Extracell Vesicles, vol.1, 2012.

A. Bobrie, M. Colombo, S. Krumeich, G. Raposo, and C. Théry, Diverse subpopulations of vesicles secreted by different intracellular mechanisms are present in exosome preparations obtained by differential ultracentrifugation, J Extracell Vesicles, vol.1, 2012.

M. A. Livshits, M. A. Livshts, E. Khomyakova, E. G. Evtushenko, V. N. Lazarev et al., Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol. Sci Rep, vol.5, p.17319, 2015.

M. C. Deregibus, F. Figliolini, D. 'antico, S. Manzini, P. M. Pasquino et al., Charge-based precipitation of extracellular vesicles, Int J Mol Med. nov, vol.38, issue.5, pp.1359-66, 2016.

K. Yakimchuk, Exosomes: Isolation and Characterization Methods and Specific Markers. Mater Methods [Internet]. 6 mars, 2019.

, Disponible sur: /method/Exosomes-Isolation-and-Characterization-Methods-and-Specific-Markers

J. D. Arroyo, J. R. Chevillet, E. M. Kroh, I. K. Ruf, C. C. Pritchard et al., Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma, Proc Natl Acad Sci, vol.108, issue.12, pp.5003-5011, 2011.

H. M. Colhoun, J. D. Otvos, M. B. Rubens, M. R. Taskinen, S. R. Underwood et al., Lipoprotein subclasses and particle sizes and their relationship with coronary artery calcification in men and women with and without type 1 diabetes, Diabetes. juin, vol.51, issue.6, pp.1949-56, 2002.

S. I. Brett, F. Lucien, C. Guo, K. C. Williams, Y. Kim et al., Immunoaffinity based methods are superior to kits for purification of prostate derived extracellular vesicles from plasma samples. The Prostate, vol.77, pp.1335-1378, 2017.

J. C. Contreras-naranjo, H. Wu, and V. M. Ugaz, Microfluidics for exosome isolation and analysis: enabling liquid biopsy for personalized medicine, Lab Chip, vol.25, issue.21, pp.3558-77, 2017.

D. D. Taylor and S. Shah, Methods of isolating extracellular vesicles impact down-stream analyses of their cargoes, Methods San Diego Calif, vol.87, pp.3-10, 2015.

J. Van-deun, P. Mestdagh, R. Sormunen, V. Cocquyt, K. Vermaelen et al., The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling, J Extracell Vesicles, vol.18, issue.2014

R. Stranska, L. Gysbrechts, J. Wouters, P. Vermeersch, K. Bloch et al., Comparison of membrane affinity-based method with size-exclusion chromatography for isolation of exosome-like vesicles from human plasma, J Transl Med. 9 janv, vol.16, issue.1, p.1, 2018.

Z. Andreu, E. Rivas, A. Sanguino-pascual, A. Lamana, M. Marazuela et al., Comparative analysis of EV isolation procedures for miRNAs detection in serum samples, J Extracell Vesicles, vol.5

P. Sharma, S. Ludwig, L. Muller, C. S. Hong, J. M. Kirkwood et al., Immunoaffinitybased isolation of melanoma cell-derived exosomes from plasma of patients with melanoma, J Extracell Vesicles, vol.7, issue.1, p.1435138, 2018.

D. Zocco and N. Zarovni, Extraction and Analysis of Extracellular Vesicle-Associated miRNAs Following Antibody-Based Extracellular Vesicle Capture from Plasma Samples, Methods Mol Biol Clifton NJ, vol.1660, pp.269-85, 2017.

E. Kowal, D. Ter-ovanesyan, A. Regev, and G. M. Church, Extracellular Vesicle Isolation and Analysis by Western Blotting, Methods Mol Biol Clifton NJ, vol.1660, pp.143-52, 2017.

R. J. Lobb, M. Becker, W. Wen, S. Wong, C. Wiegmans et al., Optimized exosome isolation protocol for cell culture supernatant and human plasma, J Extracell Vesicles, vol.17, issue.2015

C. Gardiner, D. Vizio, D. Sahoo, S. Théry, C. Witwer et al., Techniques used for the isolation and characterization of extracellular vesicles: results of a worldwide survey, J Extracell Vesicles, vol.5, p.32945, 2016.

H. Suárez, A. Gámez-valero, R. Reyes, S. López-martín, M. J. Rodríguez et al., A bead-assisted flow cytometry method for the semi-quantitative analysis of Extracellular Vesicles, Sci Rep, vol.12, issue.1, p.11271, 2017.

S. Robert, R. Lacroix, P. Poncelet, K. Harhouri, T. Bouriche et al., Highsensitivity flow cytometry provides access to standardized measurement of small-size microparticles--brief report, Arterioscler Thromb Vasc Biol. avr, vol.32, issue.4, pp.1054-1062, 2012.

W. L. Chandler, W. Yeung, and J. F. Tait, A new microparticle size calibration standard for use in measuring smaller microparticles using a new flow cytometer, J Thromb Haemost JTH. juin, vol.9, issue.6, pp.1216-1240, 2011.

D. Enderle, A. Spiel, C. M. Coticchia, E. Berghoff, R. Mueller et al., Characterization of RNA from Exosomes and Other Extracellular Vesicles Isolated by a Novel Spin Column-Based Method, Disponible sur, vol.10

A. M. Hoy and A. H. Buck, Extracellular small RNAs: what, where, why?: Figure 1, Biochem Soc Trans. 1 août, vol.40, issue.4, pp.886-90, 2012.

M. Eldh, J. Lötvall, C. Malmhäll, and K. Ekström, Importance of RNA isolation methods for analysis of exosomal RNA: evaluation of different methods, Mol Immunol. avr, vol.50, issue.4, pp.278-86, 2012.

N. J. Park, H. Zhou, D. Elashoff, B. S. Henson, D. A. Kastratovic et al., Salivary microRNA: discovery, characterization, and clinical utility for oral cancer detection, Clin Cancer Res Off J Am Assoc Cancer Res. 1 sept, vol.15, issue.17, pp.5473-5480, 2009.

M. Hanke, K. Hoefig, H. Merz, A. C. Feller, I. Kausch et al., A robust methodology to study urine microRNA as tumor marker: microRNA-126 and microRNA-182 are related to urinary bladder cancer, Urol Oncol. déc, vol.28, issue.6, pp.655-61, 2010.

D. Zubakov, A. Boersma, Y. Choi, P. F. Van-kuijk, E. Wiemer et al., MicroRNA markers for forensic body fluid identification obtained from microarray screening and quantitative RT-PCR confirmation, Int J Legal Med. mai, vol.124, issue.3, pp.217-243, 2010.

U. Irion, S. Johnston, and D. , RNA localization requires specific binding of an endosomal sorting complex, Nature. 1 févr, vol.445, issue.7127, pp.554-562, 2007.

A. B. Emerman and M. D. Blower, The RNA-binding complex ESCRT-II in Xenopus laevis eggs recognizes purine-rich sequences through its subunit, Vps25, J Biol Chem, vol.10, issue.32, pp.12593-605, 2018.

N. Kosaka, H. Iguchi, Y. Yoshioka, F. Takeshita, Y. Matsuki et al., Secretory mechanisms and intercellular transfer of microRNAs in living cells, J Biol Chem. 4 juin, vol.285, issue.23, pp.17442-52, 2010.

C. Villarroya-beltri, C. Gutiérrez-vázquez, F. Sánchez-cabo, D. Pérez-hernández, J. Vázquez et al., Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs, Nat Commun, vol.20, issue.2013

D. Koppers-lalic, M. Hackenberg, I. V. Bijnsdorp, M. Van-eijndhoven, P. Sadek et al., Nontemplated nucleotide additions distinguish the small RNA composition in cells from exosomes, Cell Rep. 25 sept, vol.8, issue.6, pp.1649-58, 2014.

L. Pigati, S. Yaddanapudi, R. Iyengar, D. Kim, S. A. Hearn et al., Selective release of microRNA species from normal and malignant mammary epithelial cells, PloS One, vol.5, issue.10, p.13515, 2010.

H. Valadi, K. Ekström, A. Bossios, M. Sjöstrand, J. J. Lee et al., Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells, Nat Cell Biol. juin, vol.9, issue.6, pp.654-663, 2007.

A. O. Batagov and I. V. Kurochkin, Exosomes secreted by human cells transport largely mRNA fragments that are enriched in the 3?-untranslated regions, Biol Direct. 7 juin, vol.8, p.12, 2013.

A. Yeri, A. Courtright, R. Reiman, E. Carlson, T. Beecroft et al., Total Extracellular Small RNA Profiles from Plasma, Saliva, and Urine of Healthy Subjects, Sci Rep, vol.17, p.44061, 2017.

T. Yuan, X. Huang, M. Woodcock, M. Du, R. Dittmar et al., Plasma extracellular RNA profiles in healthy and cancer patients, Sci Rep. 20 janv, vol.6, 2016.

M. Li, E. Zeringer, T. Barta, J. Schageman, A. Cheng et al., Analysis of the RNA content of the exosomes derived from blood serum and urine and its potential as biomarkers, Disponible sur, vol.26, p.369, 1652.

Y. Guo, K. Vickers, Y. Xiong, S. Zhao, Q. Sheng et al., Comprehensive evaluation of extracellular small RNA isolation methods from serum in high throughput sequencing, BMC Genomics, vol.18

J. E. Freedman, M. Gerstein, M. E. Rozowsky, J. Levy, D. Kitchen et al., Diverse human extracellular RNAs are widely detected in human plasma, Nat Commun, vol.7, p.11106, 2016.

X. Huang, T. Yuan, M. Tschannen, Z. Sun, H. Jacob et al., Characterization of human plasma-derived exosomal RNAs by deep sequencing, BMC Genomics, vol.14, p.319, 2013.

S. U. Umu, H. Langseth, C. Bucher-johannessen, B. Fromm, A. Keller et al., A comprehensive profile of circulating RNAs in human serum, RNA Biol, vol.15, issue.2, pp.242-50, 2018.

K. M. Danielson, R. Rubio, F. Abderazzaq, S. Das, and Y. E. Wang, High Throughput Sequencing of Extracellular RNA from Human Plasma, PloS One, vol.12, issue.1, p.164644, 2017.

H. H. Cheng, H. S. Yi, Y. Kim, E. M. Kroh, J. W. Chien et al., Plasma processing conditions substantially influence circulating microRNA biomarker levels, PloS One, vol.8, issue.6, p.64795, 2013.

J. P. Tosar, A. Cayota, E. Eitan, M. K. Halushka, and K. W. Witwer, Ribonucleic artefacts: are some extracellular RNA discoveries driven by cell culture medium components?, J Extracell Vesicles, vol.6, issue.1, p.1272832, 2017.

D. Khare, N. Goldschmidt, A. Bardugo, D. Gur-wahnon, I. Z. Ben-dov et al., Plasma microRNA profiling: Exploring better biomarkers for lymphoma surveillance, Disponible sur, vol.12, 2017.

Y. Tang, Y. Huang, L. Zheng, S. Qin, X. Xu et al., Comparison of isolation methods of exosomes and exosomal RNA from cell culture medium and serum, Int J Mol Med. sept, vol.40, issue.3, pp.834-878, 2017.

M. Ding, C. Wang, X. Lu, C. Zhang, Z. Zhou et al., Comparison of commercial exosome isolation kits for circulating exosomal microRNA profiling, Anal Bioanal Chem. juin, vol.410, issue.16, pp.3805-3819, 2018.

I. Helwa, J. Cai, M. D. Drewry, A. Zimmerman, M. B. Dinkins et al., A Comparative Study of Serum Exosome Isolation Using Differential Ultracentrifugation and Three Commercial Reagents, Disponible sur, vol.12, 2017.

G. K. Patel, M. A. Khan, H. Zubair, S. K. Srivastava, M. Khushman et al., Comparative analysis of exosome isolation methods using culture supernatant for optimum yield, purity and downstream applications, vol.9

A. M. Ainsztein, P. J. Brooks, V. G. Dugan, A. Ganguly, M. Guo et al., The NIH Extracellular RNA Communication Consortium, J Extracell Vesicles, vol.4, p.27493, 2015.

A. Galvanin, G. Dostert, L. Ayadi, V. Marchand, É. Velot et al., Diversity and heterogeneity of extracellular RNA in human plasma, Biochimie. 17 mai, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02140106

A. J. Mitchell, W. D. Gray, S. S. Hayek, Y. Ko, S. Thomas et al., Platelets confound the measurement of extracellular miRNA in archived plasma, Sci Rep, vol.13, p.32651, 2016.

B. Mateescu, E. Kowal, B. Balkom, . Van, S. Bartel et al., Obstacles and opportunities in the functional analysis of extracellular vesicle RNAan ISEV position paper, J Extracell Vesicles. 1 janv, vol.6, issue.1, p.1286095, 2017.

E. Beutler, T. Gelbart, and W. Kuhl, Interference of heparin with the polymerase chain reaction, BioTechniques. août, vol.9, issue.2, p.166, 1990.

R. Lacroix, C. Judicone, M. Mooberry, M. Boucekine, N. S. Key et al., Standardization of pre-analytical variables in plasma microparticle determination: results of the International Society on Thrombosis and Haemostasis SSC Collaborative workshop, J Thromb Haemost JTH. 2 avr, 2013.

M. Yokota, N. Tatsumi, O. Nathalang, T. Yamada, and I. Tsuda, Effects of heparin on polymerase chain reaction for blood white cells, J Clin Lab Anal, vol.13, issue.3, pp.133-173, 1999.

F. Royo, K. Schlangen, L. Palomo, E. Gonzalez, J. Conde-vancells et al., Transcriptome of extracellular vesicles released by hepatocytes, PloS One, vol.8, issue.7, p.68693, 2013.

X. Lai, M. Wang, S. D. Mcelyea, S. Sherman, M. House et al., A microRNA signature in circulating exosomes is superior to exosomal glypican-1 levels for diagnosing pancreatic cancer, Cancer Lett. 1 mai, vol.393, p.86, 2017.

H. Dejima, H. Iinuma, R. Kanaoka, N. Matsutani, and M. Kawamura, Exosomal microRNA in plasma as a non-invasive biomarker for the recurrence of non-small cell lung cancer, Oncol Lett. mars, vol.13, issue.3, pp.1256-63, 2017.

S. Maruoka, Y. Gon, S. Shikano, Y. Shintani, D. Koyama et al., Exosomal microRNAs in the serum are potential real-time biomarkers for allergic inflammation in the airway of mice, Eur Respir J. 1 sept, vol.44, p.1010, 2014.

A. Sumrin, S. Moazzam, A. A. Khan, I. Ramzan, Z. Batool et al., Exosomes as Biomarker of Cancer, Braz Arch Biol Technol [Internet], vol.61, 2018.

T. Liu, Q. Zhang, J. Zhang, C. Li, Y. Miao et al., EVmiRNA: a database of miRNA profiling in extracellular vesicles, Nucleic Acids Res. 8 janv, vol.47, issue.D1, pp.89-93, 2019.

D. Li, J. Liu, W. Wang, X. Luo, X. Zhou et al., Plasma Exosomal miRNA-122-5p and miR-300-3p as Potential Markers for Transient Ischaemic Attack in Rats. Front Aging Neurosci, vol.10, 2018.

R. Sharma, X. Huang, R. A. Brekken, and A. J. Schroit, Detection of phosphatidylserine-positive exosomes for the diagnosis of early-stage malignancies, Br J Cancer. août, vol.117, issue.4, pp.545-52, 2017.

X. Xi, T. Li, Y. Huang, J. Sun, Y. Zhu et al., RNA Biomarkers: Frontier of Precision Medicine for Cancer. Non-Coding RNA, Disponible sur, vol.20

K. Wang, H. Li, Y. Yuan, A. Etheridge, Y. Zhou et al., The complex exogenous RNA spectra in human plasma: an interface with human gut biota?, PloS One, vol.7, issue.12, p.51009, 2012.

V. Ramakrishnaiah, C. Thumann, I. Fofana, F. Habersetzer, Q. Pan et al., Exosome-mediated transmission of hepatitis C virus between human hepatoma Huh7.5 cells, Proc Natl Acad Sci, vol.110, issue.32, pp.13109-13122, 2013.

A. M. Nour and Y. Modis, Endosomal vesicles as vehicles for viral genomes, Trends Cell Biol. août, vol.24, issue.8, pp.449-54, 2014.

J. M. Smit, B. Moesker, I. Rodenhuis-zybert, and J. Wilschut, Flavivirus Cell Entry and Membrane Fusion, Viruses. 22 févr, vol.3, issue.2, pp.160-71, 2011.

R. Hamel, O. Dejarnac, S. Wichit, P. Ekchariyawat, A. Neyret et al., Biology of Zika Virus Infection in Human Skin Cells, J Virol. 17 juin, vol.89, issue.17, pp.8880-96, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01228435

A. Longatti, The Dual Role of Exosomes in Hepatitis A and C Virus Transmission and Viral Immune Activation, Viruses. 17 déc, vol.7, issue.12, pp.6707-6722, 2015.

M. R. Anderson, F. Kashanchi, and S. Jacobson, Exosomes in Viral Disease, Neurotherapeutics. juill, vol.13, issue.3, pp.535-581, 2016.

J. D. Arroyo, J. R. Chevillet, E. M. Kroh, I. K. Ruf, C. C. Pritchard et al., Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma, Proc Natl Acad Sci, vol.108, issue.12, pp.5003-5011, 2011.

M. P. Kowalski and T. Krude, Functional roles of non-coding Y RNAs, Int J Biochem Cell Biol. sept, vol.66, pp.20-29, 2015.

J. M. Dhahbi, S. R. Spindler, H. Atamna, D. Boffelli, P. Mote et al., 5?-YRNA fragments derived by processing of transcripts from specific YRNA genes and pseudogenes are abundant in human serum and plasma, Physiol Genomics, vol.45, issue.21, pp.990-998, 2013.

S. Ninomiya, M. Kawano, T. Abe, T. Ishikawa, M. Takahashi et al., Potential Small Guide RNAs for tRNase ZL from Human Plasma, Peripheral Blood Mononuclear Cells, and Cultured Cell Lines, Disponible sur, vol.10

C. P. Christov, E. Trivier, and T. Krude, Noncoding human Y RNAs are overexpressed in tumours and required for cell proliferation, Br J Cancer. 11 mars, vol.98, issue.5, pp.981-989, 2008.

M. B. Hoagland, E. B. Keller, and P. C. Zamecnik, Enzymatic carboxyl activation of amino acids, J Biol Chem. janv, vol.218, issue.1, pp.345-58, 1956.

M. B. Hoagland, M. L. Stephenson, J. F. Scott, L. I. Hecht, and P. C. Zamecnik, A soluble ribonucleic acid intermediate in protein synthesis, J Biol Chem. mars, vol.231, issue.1, pp.241-57, 1958.

M. B. Hoagland, P. C. Zamecnik, and M. L. Stephenson, Intermediate reactions in protein biosynthesis, Biochim Biophys Acta, vol.1000, pp.106-113, 1957.

U. L. Rajbhandary and C. Köhrer, Early days of tRNA research: discovery, function, purification and sequence analysis, J Biosci, vol.31, issue.4, pp.439-51, 2006.

R. W. Holley, Structure of an alanine transfer ribonucleic acid, JAMA, vol.22, issue.8, pp.868-71, 1965.

J. Shepherd and M. Ibba, Bacterial transfer RNAs, FEMS Microbiol Rev. mai, vol.39, issue.3, pp.280-300, 2015.

H. Hori, C. Tomikawa, A. Hirata, Y. Toh, K. Tomita et al., Transfer RNA Synthesis and Regulation, p.eLS

D. Sur,

M. J. Fournier and H. Ozeki, Structure and organization of the transfer ribonucleic acid genes of Escherichia coli K-12, Microbiol Rev. déc, vol.49, issue.4, pp.379-97, 1985.

M. P. Deutscher, J. J. Lin, and J. A. Evans, Transfer RNA metabolism in Escherichia coli cells deficient in tRNA nucleotidyltransferase, J Mol Biol. 25 déc, vol.117, issue.4, pp.1081-94, 1977.

E. Lizano, M. Scheibe, C. Rammelt, H. Betat, and M. Mörl, A comparative analysis of CCAadding enzymes from human and E. coli: differences in CCA addition and tRNA 3'-end repair, Biochimie. mai, vol.90, issue.5, pp.762-72, 2008.

H. D. Robertson, R. E. Webster, and N. D. Zinder, Purification and properties of ribonuclease III from Escherichia coli, J Biol Chem. 10 janv, vol.243, issue.1, pp.82-91, 1968.

K. J. Mcdowall, S. Lin-chao, and S. N. Cohen, A+U content rather than a particular nucleotide order determines the specificity of RNase E cleavage, J Biol Chem. 8 avr, vol.269, issue.14, pp.10790-10796, 1994.

A. W. Nicholson, Function, mechanism and regulation of bacterial ribonucleases, FEMS Microbiol Rev. juin, vol.23, issue.3, pp.371-90, 1999.

P. Gegenheimer and D. Apirion, Processing of procaryotic ribonucleic acid, Microbiol Rev. déc, vol.45, issue.4, pp.502-543, 1981.

G. Plautz and D. Apirion, Processing of RNA in Escherichia coli is limited in the absence of ribonuclease III, ribonuclease E and ribonuclease P, J Mol Biol. 15 juill, vol.149, issue.4, pp.813-822, 1981.

H. Cudny and M. P. Deutscher, Apparent involvement of ribonuclease D in the 3' processing of tRNA precursors, Proc Natl Acad Sci U S A. févr, vol.77, issue.2, pp.837-878, 1980.

R. K. Ghosh and M. P. Deutscher, Purification of potential 3' processing nucleases using synthetic tRNA precursors, Nucleic Acids Res. oct, vol.5, issue.10, pp.3831-3873, 1978.

S. Altman, Biosynthesis of transfer RNA in Escherichia coli, Cell. janv, vol.4, issue.1, pp.21-30, 1975.

L. A. Kirsebom, RNase P RNA-mediated catalysis, Biochem Soc Trans, vol.30, pp.1153-1161, 2002.

M. E. Harris and E. L. Christian, Recent insights into the structure and function of the ribonucleoprotein enzyme ribonuclease P, Curr Opin Struct Biol. juin, vol.13, issue.3, pp.325-358, 2003.

A. V. Kazantsev and N. R. Pace, Bacterial RNase P: a new view of an ancient enzyme, Nat Rev Microbiol, vol.4, issue.10, pp.729-769, 2006.

A. Torres-larios, K. K. Swinger, T. Pan, and A. Mondragón, Structure of ribonuclease P--a universal ribozyme, Curr Opin Struct Biol. juin, vol.16, issue.3, pp.327-362, 2006.

B. C. Stark, R. Kole, E. J. Bowman, and S. Altman, Ribonuclease P: an enzyme with an essential RNA component, Proc Natl Acad Sci U S A. août, vol.75, issue.8, pp.3717-3738, 1978.

C. Guthrie and R. Atchison, Biochemical Characterization of RNase P: A tRNA Processing Activity with Protein and RNA Components, Cold Spring Harb Monogr Arch. 1 janv, vol.09, issue.0, pp.83-97, 1980.

S. C. Walker and D. R. Engelke, Ribonuclease P: the evolution of an ancient RNA enzyme, Crit Rev Biochem Mol Biol. avr, vol.41, issue.2, pp.77-102, 2006.

J. K. Smith, J. Hsieh, and C. A. Fierke, Importance of RNA-protein interactions in bacterial ribonuclease P structure and catalysis, Biopolymers. 5 déc, vol.87, issue.5-6, pp.329-367, 2007.

U. Guenther, L. E. Yandek, C. N. Niland, F. E. Campbell, D. Anderson et al., Hidden specificity in an apparently nonspecific RNA-binding protein, Nature, vol.502, issue.7471, pp.385-393, 2013.

H. Grosjean and . Dna, RNA Modification Enzymes : Structure, Mechanism, Function and Evolution, 2009.

Y. Motorin and M. Helm, tRNA stabilization by modified nucleotides, Biochemistry. 22 juin, vol.49, issue.24, pp.4934-4978, 2010.

B. El-yacoubi, M. Bailly, and V. De-crécy-lagard, Biosynthesis and function of posttranscriptional modifications of transfer RNAs, Annu Rev Genet, vol.46, pp.69-95, 2012.

G. R. Björk, J. U. Ericson, C. E. Gustafsson, T. G. Hagervall, Y. H. Jönsson et al., Transfer RNA modification, Annu Rev Biochem, vol.56, pp.263-87, 1987.

B. Addepalli and P. A. Limbach, Pseudouridine in the Anticodon of Escherichia coli tRNATyr(Q?A) Is Catalyzed by the Dual Specificity Enzyme RluF, J Biol Chem, vol.291, issue.42, pp.22327-22364, 2016.

S. Gehrig, M. Eberle, F. Botschen, K. Rimbach, F. Eberle et al., Identification of modifications in microbial, native tRNA that suppress immunostimulatory activity, J Exp Med. 13 févr, vol.209, issue.2, pp.225-258, 2012.

K. Rimbach, S. Kaiser, M. Helm, A. H. Dalpke, and T. Eigenbrod, 2'-O-Methylation within Bacterial RNA Acts as Suppressor of TLR7/TLR8 Activation in Human Innate Immune Cells, J Innate Immun, vol.7, issue.5, pp.482-93, 2015.

M. Ibba and D. Soll, Aminoacyl-tRNA synthesis, Annu Rev Biochem, vol.69, pp.617-50, 2000.

N. M. Reynolds, B. A. Lazazzera, and M. Ibba, Cellular mechanisms that control mistranslation, Nat Rev Microbiol. déc, vol.8, issue.12, pp.849-56, 2010.

L. Ayadi, A. Galvanin, F. Pichot, V. Marchand, and Y. Motorin, RNA ribose methylation (2'-Omethylation): Occurrence, biosynthesis and biological functions, Biochim Biophys Acta Gene Regul Mech. mars, vol.1862, issue.3, pp.253-69, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01969343

M. A. Machnicka, K. Milanowska, O. Oglou, O. Purta, E. Kurkowska et al., MODOMICS: a database of RNA modification pathways--2013 update, Nucleic Acids Res. janv, vol.41, pp.262-267, 2013.

H. L. Schubert, R. M. Blumenthal, and X. Cheng, Many paths to methyltransfer: a chronicle of convergence, Trends Biochem Sci. juin, vol.28, issue.6, pp.329-364, 2003.

S. Boschi-muller and Y. Motorin, Chemistry enters nucleic acids biology: enzymatic mechanisms of RNA modification, Biochem Biokhimiia. déc, vol.78, issue.13, pp.1392-404, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01467256

P. Z. Kozbial and A. R. Mushegian, Natural history of S-adenosylmethionine-binding proteins, BMC Struct Biol, vol.5, p.19, 2005.

H. Hori, Transfer RNA methyltransferases with a SpoU-TrmD (SPOUT) fold and their modified nucleosides in tRNA, Biomolecules. 28 févr, vol.7, issue.1, p.23, 2017.

K. Watanabe, O. Nureki, S. Fukai, Y. Endo, and H. Hori, Functional Categorization of the Conserved Basic Amino Acid Residues in TrmH (tRNA (Gm18) Methyltansferase) Enzymes, J Biol Chem, vol.281, issue.45, pp.34630-34639, 2006.

K. Watanabe, O. Nureki, S. Fukai, R. Ishii, H. Okamoto et al., Roles of conserved amino acid sequence motifs in the SpoU (TrmH) RNA methyltransferase family, J Biol Chem. 18 mars, vol.280, issue.11, pp.10368-77, 2005.

B. C. Persson, G. Jäger, and C. Gustafsson, The spoU gene of Escherichia coli, the fourth gene of the spoT operon, is essential for tRNA (Gm18) 2'-O-methyltransferase activity, Nucleic Acids Res, vol.25, issue.20, pp.4093-4100, 1997.

I. Kumagai, K. Watanabe, and T. Oshima, A thermostable tRNA (guanosine-2')-methyltransferase from Thermus thermophilus HB27 and the effect of ribose methylation on the conformational stability of tRNA, J Biol Chem. 10 juill, vol.257, issue.13, pp.7388-95, 1982.

H. Hori, T. Suzuki, K. Sugawara, Y. Inoue, T. Shibata et al., Identification and characterization of tRNA (Gm18) methyltransferase from Thermus thermophilus HB8: domain structure and conserved amino acid sequence motifs, Genes Cells Devoted Mol Cell Mech. mars, vol.7, issue.3, pp.259-72, 2002.

O. Nureki, K. Watanabe, S. Fukai, R. Ishii, Y. Endo et al., Deep knot structure for construction of active site and cofactor binding site of tRNA modification enzyme, Struct Lond Engl, vol.12, issue.4, pp.593-602, 1993.

E. Purta, F. Van-vliet, K. L. Tkaczuk, S. Dunin-horkawicz, H. Mori et al., The yfhQ gene of Escherichia coli encodes a tRNA:Cm32/Um32 methyltransferase, BMC Mol Biol. 18 juill, vol.7, p.23, 2006.

J. Somme, B. Van-laer, M. Roovers, J. Steyaert, W. Versées et al., Characterization of two homologous 2?-O-methyltransferases showing different specificities for their tRNA substrates, RNA. août, vol.20, issue.8, pp.1257-71, 2014.

R. Liu, T. Long, M. Zhou, X. Zhou, and E. Wang, tRNA recognition by a bacterial tRNA Xm32 modification enzyme from the SPOUT methyltransferase superfamily, Nucleic Acids Res. 3 sept, vol.43, issue.15, pp.7489-503, 2015.

A. Benítez-páez, M. Villarroya, S. Douthwaite, T. Gabaldón, and M. Armengod, YibK is the 2'-O-methyltransferase TrmL that modifies the wobble nucleotide in Escherichia coli tRNA(Leu) isoacceptors. RNA N Y N, vol.16, pp.2131-2174, 2010.

M. Armengod, I. Moukadiri, S. Prado, R. Ruiz-partida, A. Benítez-páez et al., Enzymology of tRNA modification in the bacterial MnmEG pathway, Biochimie. juill, vol.94, issue.7, pp.1510-1530, 2012.

G. Kawai, H. Ue, M. Yasuda, K. Sakamoto, T. Hashizume et al., Relation between functions and conformational characteristics of modified nucleosides found in tRNAs, Nucleic Acids Symp Ser, issue.25, pp.49-50, 1991.

P. Prusiner, N. Yathindra, and M. Sundaralingam, Effect of ribose O(2')-methylation on the conformation of nucleosides and nucleotides, Biochim Biophys Acta, vol.366, issue.2, pp.115-138, 1974.

B. G. Lane and T. Tamaoki, Studies of the chain termini and alkali-stable dinucleotide sequences in 16 s and 28 s ribosomal RNA from L cells, J Mol Biol. 28 juill, vol.27, issue.2, pp.335-383, 1967.

A. R. Trim and J. E. Parker, Preparation, purification and analyses of thirteen alkali-stable dinucleotides from yeast ribonucleic acid, Biochem J. févr, vol.116, issue.4, pp.589-98, 1970.

L. Endres, P. C. Dedon, and T. J. Begley, Codon-biased translation can be regulated by wobblebase tRNA modification systems during cellular stress responses, RNA Biol. 18 avr, vol.12, issue.6, pp.603-617, 2015.

H. Huang and A. K. Hopper, Multiple Layers of Stress-Induced Regulation in tRNA Biology, Disponible sur, vol.6, 2016.

K. Pluta, O. Lefebvre, N. C. Martin, W. J. Smagowicz, D. R. Stanford et al., Maf1p, a negative effector of RNA polymerase III in Saccharomyces cerevisiae, Mol Cell Biol. août, vol.21, issue.15, pp.5031-5071, 2001.

A. J. Oler and B. R. Cairns, PP4 dephosphorylates Maf1 to couple multiple stress conditions to RNA polymerase III repression, EMBO J. 21 mars, vol.31, issue.6, pp.1440-52, 2012.

A. Vannini, R. Ringel, A. G. Kusser, O. Berninghausen, G. A. Kassavetis et al., Molecular basis of RNA polymerase III transcription repression by Maf1, Cell, vol.143, issue.1, pp.59-70, 2010.

D. Oficjalska-pham, O. Harismendy, W. J. Smagowicz, A. Gonzalez-de-peredo, M. Boguta et al., General repression of RNA polymerase III transcription is triggered by protein phosphatase type 2A-mediated dephosphorylation of Maf1, Mol Cell. 9 juin, vol.22, issue.5, pp.623-655, 2006.

D. N. Roberts, B. Wilson, J. T. Huff, A. J. Stewart, and B. R. Cairns, Dephosphorylation and genomewide association of Maf1 with Pol III-transcribed genes during repression, Mol Cell. 9 juin, vol.22, issue.5, pp.633-677, 2006.

R. Upadhya, J. Lee, and I. M. Willis, Maf1 is an essential mediator of diverse signals that repress RNA polymerase III transcription, Mol Cell. déc, vol.10, issue.6, pp.1489-94, 2002.

R. V. Intine, A. L. Sakulich, S. B. Koduru, Y. Huang, E. Pierstorff et al., Control of transfer RNA maturation by phosphorylation of the human La antigen on serine 366, Mol Cell. août, vol.6, issue.2, pp.339-387, 2000.

R. V. Intine, S. A. Tenenbaum, A. L. Sakulich, J. D. Keene, and R. J. Maraia, Differential phosphorylation and subcellular localization of La RNPs associated with precursor tRNAs and translation-related mRNAs, Mol Cell, vol.12, issue.5, pp.1301-1308, 2003.

D. Foretek, J. Wu, A. K. Hopper, and M. Boguta, Control of Saccharomyces cerevisiae pre-tRNA processing by environmental conditions, RNA N Y N. mars, vol.22, issue.3, pp.339-388, 2016.

N. Netzer, J. M. Goodenbour, A. David, K. A. Dittmar, R. B. Jones et al., Innate immune and chemically triggered oxidative stress modifies translational fidelity, Nature, vol.462, issue.7272, pp.522-528, 2009.

E. Wiltrout, J. M. Goodenbour, M. Fréchin, and T. Pan, Misacylation of tRNA with methionine in Saccharomyces cerevisiae, Nucleic Acids Res, vol.40, issue.20, pp.10494-506, 2012.

G. Kaufmann, Anticodon nucleases, Trends Biochem Sci. févr, vol.25, issue.2, pp.70-74, 2000.

S. Kirchner and Z. Ignatova, Emerging roles of tRNA in adaptive translation, signalling dynamics and disease, Nat Rev Genet. févr, vol.16, issue.2, pp.98-112, 2015.

D. M. Thompson, C. Lu, P. J. Green, R. Parker, and . Rna-n-y-n, tRNA cleavage is a conserved response to oxidative stress in eukaryotes, vol.14, pp.2095-103, 2008.

H. Masaki and T. Ogawa, The modes of action of colicins E5 and D, and related cytotoxic tRNases, Biochimie. juin, vol.84, issue.5-6, pp.433-441, 2002.

P. Anderson and P. Ivanov, tRNA fragments in human health and disease, FEBS Lett, vol.588, issue.23, pp.4297-304, 2014.

J. Gebetsberger and N. Polacek, Slicing tRNAs to boost functional ncRNA diversity, RNA Biol. déc, vol.10, issue.12, pp.1798-806, 2013.

D. M. Thompson and R. Parker, Stressing out over tRNA cleavage, Cell. 23 juill, vol.138, issue.2, pp.215-224, 2009.

H. Fu, J. Feng, Q. Liu, F. Sun, Y. Tie et al., Stress induces tRNA cleavage by angiogenin in mammalian cells, FEBS Lett. 22 janv, vol.583, issue.2, pp.437-479, 2009.

P. Ivanov, M. M. Emara, J. Villen, S. P. Gygi, and P. Anderson, Angiogenin-induced tRNA fragments inhibit translation initiation, Mol Cell. 19 août, vol.43, issue.4, pp.613-636, 2011.

P. Ivanov, E. O'day, M. M. Emara, G. Wagner, J. Lieberman et al., G-quadruplex structures contribute to the neuroprotective effects of angiogenin-induced tRNA fragments, Proc Natl Acad Sci, vol.111, issue.51, pp.18201-18207, 2014.

M. Saikia, R. Jobava, M. Parisien, A. Putnam, D. Krokowski et al., Angiogenincleaved tRNA halves interact with cytochrome c, protecting cells from apoptosis during osmotic stress, Mol Cell Biol. juill, vol.34, issue.13, pp.2450-63, 2014.

J. R. Zamudio, T. J. Kelly, and P. A. Sharp, Argonaute-bound small RNAs from promoter-proximal RNA polymerase II, Cell. 27 févr, vol.156, issue.5, pp.920-954, 2014.

R. L. Maute, C. Schneider, P. Sumazin, A. Holmes, A. Califano et al., tRNA-derived microRNA modulates proliferation and the DNA damage response and is down-regulated in B cell lymphoma, Proc Natl Acad Sci, vol.110, issue.4, pp.1404-1413, 2013.

J. Gebetsberger, M. Zywicki, A. Künzi, and N. Polacek, tRNA-derived fragments target the ribosome and function as regulatory non-coding RNA in Haloferax volcanii, Archaea Vanc BC, p.260909, 2012.

M. A. Sørensen, A. O. Fehler, L. Svenningsen, and S. , Transfer RNA instability as a stress response in Escherichia coli: Rapid dynamics of the tRNA pool as a function of demand, RNA Biol. 9 mai, vol.15, issue.4-5, pp.586-93, 2018.

K. R. Noon, R. Guymon, P. F. Crain, J. A. Mccloskey, M. Thomm et al., Influence of Temperature on tRNA Modification in Archaea: Methanococcoides burtonii (Optimum Growth Temperature [Topt], 23°C) and Stetteria hydrogenophila (Topt, 95°C), J Bacteriol. sept, vol.185, issue.18, pp.5483-90, 2003.

S. M. Kinghorn, C. P. O'byrne, I. R. Booth, and I. Stansfield, Physiological analysis of the role of truB in Escherichia coli: a role for tRNA modification in extreme temperature resistance. Microbiol Read Engl, vol.148, pp.3511-3531, 2002.

M. Murata, H. Fujimoto, K. Nishimura, K. Charoensuk, H. Nagamitsu et al., Molecular strategy for survival at a critical high temperature in Eschierichia coli, PloS One, vol.6, issue.6, p.20063, 2011.

V. Emilsson, A. K. Näslund, and C. G. Kurland, Thiolation of transfer RNA in Escherichia coli varies with growth rate, Nucleic Acids Res. 11 sept, vol.20, issue.17, pp.4499-505, 1992.

K. M. Thompson and S. Gottesman, The MiaA tRNA modification enzyme is necessary for robust RpoS expression in Escherichia coli, J Bacteriol. févr, vol.196, issue.4, pp.754-61, 2014.

L. Han, Y. Kon, and E. M. Phizicky, Functional importance of ?38 and ?39 in distinct tRNAs, amplified for tRNAGln(UUG) by unexpected temperature sensitivity of the s2U modification in yeast, RNA N Y N. févr, vol.21, issue.2, pp.188-201, 2015.

M. A. Preston, S. Silva, Y. Kon, and E. M. Phizicky, tRNAHis 5-methylcytidine levels increase in response to several growth arrest conditions in Saccharomyces cerevisiae, RNA N Y N. févr, vol.19, issue.2, pp.243-56, 2013.

F. Alings, L. P. Sarin, C. Fufezan, H. Drexler, and S. A. Leidel, An evolutionary approach uncovers a diverse response of tRNA 2-thiolation to elevated temperatures in yeast, RNA N Y N. févr, vol.21, issue.2, pp.202-214, 2015.

J. R. Damon, D. Pincus, and H. L. Ploegh, tRNA thiolation links translation to stress responses in Saccharomyces cerevisiae, Mol Biol Cell. 15 janv, vol.26, issue.2, pp.270-82, 2015.

C. Chan, M. Dyavaiah, M. S. Demott, K. Taghizadeh, P. C. Dedon et al., A quantitative systems approach reveals dynamic control of tRNA modifications during cellular stress, PLoS Genet. 16 déc, vol.6, issue.12, p.1001247, 2010.

C. Gu, T. J. Begley, and P. C. Dedon, tRNA modifications regulate translation during cellular stress, FEBS Lett, vol.588, issue.23, pp.4287-96, 2014.

D. Dominissini, S. Moshitch-moshkovitz, S. Schwartz, M. Salmon-divon, L. Ungar et al., Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq, Nature. 29 avr, vol.485, issue.7397, pp.201-207, 2012.

J. E. Squires, H. R. Patel, M. Nousch, T. Sibbritt, D. T. Humphreys et al., Widespread occurrence of 5-methylcytosine in human coding and non-coding RNA, Nucleic Acids Res. juin, vol.40, issue.11, pp.5023-5056, 2012.

T. M. Carlile, M. F. Rojas-duran, B. Zinshteyn, H. Shin, K. M. Bartoli et al., Pseudouridine profiling reveals regulated mRNA pseudouridylation in yeast and human cells, Nature, vol.515, issue.7525, pp.143-149, 2014.

S. Edelheit, S. Schwartz, M. R. Mumbach, O. Wurtzel, and R. Sorek, Transcriptome-wide mapping of 5-methylcytidine RNA modifications in bacteria, archaea, and yeast reveals m5C within archaeal mRNAs, PLoS Genet. juin, vol.9, issue.6, p.1003602, 2013.

K. D. Meyer, Y. Saletore, P. Zumbo, O. Elemento, C. E. Mason et al., Comprehensive analysis of mRNA methylation reveals enrichment in 3' UTRs and near stop codons, Cell. 22 juin, vol.149, issue.7, pp.1635-1681, 2012.

A. V. Grozhik and S. R. Jaffrey, Distinguishing RNA modifications from noise in epitranscriptome maps, Nat Chem Biol, vol.14, issue.3, pp.215-240, 2018.

P. Ryvkin, Y. Y. Leung, I. M. Silverman, M. Childress, O. Valladares et al., HAMR: high-throughput annotation of modified ribonucleotides, RNA N Y N. déc, vol.19, issue.12, pp.1684-92, 2013.

Y. Motorin and M. Helm, Methods for RNA Modification Mapping Using Deep Sequencing: Established and New Emerging Technologies, Genes. 9 janv, vol.10, issue.1, 2019.

A. E. Cozen, E. Quartley, A. D. Holmes, E. Hrabeta-robinson, E. M. Phizicky et al., ARMseq: AlkB-facilitated RNA methylation sequencing reveals a complex landscape of modified tRNA fragments, Nat Methods. sept, vol.12, issue.9, pp.879-84, 2015.

G. Zheng, Y. Qin, W. C. Clark, Q. Dai, C. Yi et al., Efficient and quantitative highthroughput tRNA sequencing, Nat Methods. sept, vol.12, issue.9, pp.835-842, 2015.

Q. Dai, G. Zheng, M. H. Schwartz, W. C. Clark, and T. Pan, Selective Enzymatic Demethylation of N2 ,N2 -Dimethylguanosine in RNA and Its Application in High-Throughput tRNA Sequencing, Angew Chem Int Ed Engl, vol.24, issue.18, pp.5017-5037, 2017.

M. Schaefer, T. Pollex, K. Hanna, and F. Lyko, RNA cytosine methylation analysis by bisulfite sequencing, Nucleic Acids Res. févr, vol.37, issue.2, p.12, 2009.

T. M. Carlile, M. F. Rojas-duran, and W. V. Gilbert, Pseudo-Seq: Genome-Wide Detection of Pseudouridine Modifications in RNA, Methods Enzymol, vol.560, pp.219-264, 2015.

T. M. Carlile, M. F. Rojas-duran, and W. V. Gilbert, Transcriptome-Wide Identification of Pseudouridine Modifications Using Pseudo-seq, Curr Protoc Mol Biol, 2015.

S. Schwartz, D. A. Bernstein, M. R. Mumbach, M. Jovanovic, R. H. Herbst et al., Transcriptome-wide mapping reveals widespread dynamic-regulated pseudouridylation of ncRNA and mRNA, Cell. 25 sept, vol.159, issue.1, pp.148-62, 2014.

A. F. Lovejoy, D. P. Riordan, and P. O. Brown, Transcriptome-wide mapping of pseudouridines: pseudouridine synthases modify specific mRNAs in S. cerevisiae, PloS One, vol.9, issue.10, p.110799, 2014.

M. A. Nakamoto, A. F. Lovejoy, A. M. Cygan, and J. C. Boothroyd, mRNA pseudouridylation affects RNA metabolism in the parasite Toxoplasma gondii, RNA N Y N, vol.23, issue.12, pp.1834-1883, 2017.

V. Marchand, L. Ayadi, F. Ernst, J. Hertler, V. Bourguignon-igel et al., AlkAniline-Seq: Profiling of m7 G and m3 C RNA Modifications at Single Nucleotide Resolution, Angew Chem Int Ed Engl. 17 déc, vol.57, issue.51, pp.16785-90, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01928435

B. E. Maden, Mapping 2'-O-methyl groups in ribosomal RNA. Methods San Diego Calif, vol.25, pp.374-82, 2001.

D. Incarnato, F. Anselmi, E. Morandi, F. Neri, M. Maldotti et al., High-throughput single-base resolution mapping of RNA 2?-O-methylated residues, Nucleic Acids Res, vol.17, issue.3, pp.1433-1474, 2017.

Z. Dong, P. Shao, L. Diao, H. Zhou, C. Yu et al., RTL-P: a sensitive approach for detecting sites of 2'-O-methylation in RNA molecules, Nucleic Acids Res, vol.40, issue.20, p.157, 2012.

J. Aschenbrenner and A. Marx, Direct and site-specific quantification of RNA 2'-Omethylation by PCR with an engineered DNA polymerase, Nucleic Acids Res. 05, vol.44, issue.8, pp.3495-502, 2016.

Q. Dai, S. Moshitch-moshkovitz, D. Han, N. Kol, N. Amariglio et al., Nm-seq maps 2'-O-methylation sites in human mRNA with base precision, Nat Methods. juill, vol.14, issue.7, pp.695-703, 2017.

Y. Zhu, S. P. Pirnie, and G. G. Carmichael, High-throughput and site-specific identification of 2'-O-methylation sites using ribose oxidation sequencing (RibOxi-seq), RNA N Y N, vol.23, issue.8, pp.1303-1317, 2017.

U. Birkedal, M. Christensen-dalsgaard, N. Krogh, R. Sabarinathan, J. Gorodkin et al., Profiling of ribose methylations in RNA by high-throughput sequencing, Angew Chem Int Ed Engl. 7 janv, vol.54, issue.2, pp.451-456, 2015.

V. Marchand, F. Blanloeil-oillo, M. Helm, and Y. Motorin, Illumina-based RiboMethSeq approach for mapping of 2'-O-Me residues in RNA, Nucleic Acids Res. 14 juin, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01452312

V. Marchand, F. Pichot, K. Thüring, L. Ayadi, I. Freund et al., Next-Generation Sequencing-Based RiboMethSeq Protocol for Analysis of tRNA 2?-O-Methylation
URL : https://hal.archives-ouvertes.fr/hal-01799272

. Biomolecules, Disponible sur, vol.7, 2017.

S. Schwartz and Y. Motorin, Next-generation sequencing technologies for detection of modified nucleotides in RNAs, RNA Biol, vol.14, issue.9, pp.1124-1161, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01799254

J. Dabney and M. Meyer, Length and GC-biases during sequencing library amplification: A comparison of various polymerase-buffer systems with ancient and modern DNA sequencing libraries, BioTechniques. 1 févr, vol.52, issue.2, pp.87-94, 2012.

I. Abnizova, . Boekhorst-r-te, and Y. L. Orlov, Computational Errors and Biases in Short Read Next Generation Sequencing, J Proteomics Bioinform, vol.10, issue.1, 2017.

S. Schwartz, R. Oren, and G. Ast, Detection and Removal of Biases in the Analysis of Next-Generation Sequencing Reads, PLOS ONE. 31 janv, vol.6, issue.1, p.16685, 2011.

S. Kellner, A. Ochel, K. Thüring, F. Spenkuch, J. Neumann et al., Absolute and relative quantification of RNA modifications via biosynthetic isotopomers, Nucleic Acids Res, vol.42, issue.18, p.142, 2014.

D. Su, C. Chan, C. Gu, K. S. Lim, Y. H. Chionh et al., Quantitative analysis of ribonucleoside modifications in tRNA by HPLC-coupled mass spectrometry, Nat Protoc. avr, vol.9, issue.4, pp.828-869, 2014.

A. Galvanin, L. Ayadi, M. Helm, Y. Motorin, and V. Marchand, Mapping and Quantification of tRNA 2'-O-Methylation by RiboMethSeq, Methods Mol Biol Clifton NJ, vol.1870, pp.273-95, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01957102

M. A. Kohanski, D. J. Dwyer, and J. J. Collins, How antibiotics kill bacteria: from targets to networks, Nat Rev Microbiol. juin, vol.8, issue.6, pp.423-458, 2010.

M. G. Abouhaidar and I. G. Ivanov, Non-Enzymatic RNA Hydrolysis Promoted by the Combined Catalytic Activity of Buffers and Magnesium Ions, Z Für Naturforschung C. 1 août, vol.54, issue.7-8, pp.542-550, 1999.

D. Jacob, K. Thüring, A. Galliot, V. Marchand, A. Galvanin et al., Absolute Quantification of Noncoding RNA by Microscale Thermophoresis, Angew Chem Int Ed Engl. 20 mars, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02078064

T. Schubert and G. Längst, Studying epigenetic interactions using MicroScale Thermophoresis (MST), vol.2, pp.370-380, 2015.

D. Sur,

R. Hauenschild, L. Tserovski, K. Schmid, K. Thüring, M. Winz et al., The reverse transcription signature of N-1-methyladenosine in RNA-Seq is sequence dependent, Nucleic Acids Res, vol.43, issue.20, pp.9950-64, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01451907

Y. Pang, R. Abo, S. S. Levine, and P. C. Dedon, Diverse cell stresses induce unique patterns of tRNA up-and down-regulation: tRNA-seq for quantifying changes in tRNA copy number, Nucleic Acids Res. 16 déc, vol.42, issue.22, p.170, 2014.

S. Jung, T. Von-thülen, V. Laukemper, S. Pigisch, D. Hangel et al., A single naturally occurring 2'-O-methylation converts a TLR7-and TLR8-activating RNA into a TLR8-specific ligand, PloS One, vol.10, issue.3, p.120498, 2015.

A. D. Judge, G. Bola, A. Lee, and I. Maclachlan, Design of noninflammatory synthetic siRNA mediating potent gene silencing in vivo, Mol Ther J Am Soc Gene Ther. mars, vol.13, issue.3, pp.494-505, 2006.

M. Robbins, A. Judge, L. Liang, K. Mcclintock, E. Yaworski et al., 2'-O-methylmodified RNAs act as TLR7 antagonists, Mol Ther J Am Soc Gene Ther. sept, vol.15, issue.9, pp.1663-1672, 2007.

S. Kaiser, K. Rimbach, T. Eigenbrod, A. H. Dalpke, and M. Helm, A modified dinucleotide motif specifies tRNA recognition by TLR7, RNA. sept, vol.20, issue.9, pp.1351-1356, 2014.

B. B. Finlay and G. Mcfadden, Anti-Immunology: Evasion of the Host Immune System by Bacterial and Viral Pathogens, Cell. 24 févr, vol.124, issue.4, pp.767-82, 2006.

N. Gratz, H. Hartweger, U. Matt, F. Kratochvill, J. M. Sigel et al., Type I interferon production induced by Streptococcus pyogenes-derived nucleic acids is required for host protection, PLoS Pathog. mai, vol.7, issue.5, p.1001345, 2011.

I. Freund, D. K. Buhl, S. Boutin, A. Kotter, F. Pichot et al., 2'-O-methylation within prokaryotic and eukaryotic tRNA inhibits innate immune activation by endosomal Toll-like receptors but does not affect recognition of whole organisms, RNA N Y N. juill, vol.25, issue.7, pp.869-80, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02113259

L. E. Sander, M. J. Davis, M. V. Boekschoten, D. Amsen, C. C. Dascher et al., Detection of prokaryotic mRNA signifies microbial viability and promotes immunity, Nature. 22 mai, vol.474, issue.7351, pp.385-394, 2011.

T. Vierbuchen, C. Bang, H. Rosigkeit, R. A. Schmitz, and H. Heine, The Human-Associated Archaeon Methanosphaera stadtmanae Is Recognized through Its RNA and Induces TLR8-Dependent NLRP3 Inflammasome Activation, Front Immunol, vol.8, p.1535, 2017.

Z. Feng, L. Hensley, K. L. Mcknight, F. Hu, V. Madden et al., A pathogenic picornavirus acquires an envelope by hijacking cellular membranes, Nature. 18 avr, vol.496, issue.7445, pp.367-71, 2013.

S. Chapuy-regaud, M. Dubois, C. Plisson-chastang, T. Bonnefois, S. Lhomme et al., Characterization of the lipid envelope of exosome encapsulated HEV particles protected from the immune response, Biochimie, vol.141, pp.70-79, 2017.

M. A. Ansari, V. V. Singh, S. Dutta, M. V. Veettil, D. Dutta et al., Constitutive interferon-inducible protein 16-inflammasome activation during Epstein-Barr virus latency I, II, and III in B and epithelial cells, J Virol. août, vol.87, issue.15, pp.8606-8629, 2013.

L. Abrami, L. Brandi, M. Moayeri, M. J. Brown, B. A. Krantz et al., Hijacking multivesicular bodies enables long-term and exosome-mediated long-distance action of anthrax toxin, Cell Rep, vol.5, issue.4, pp.986-96, 2013.

A. Laganà, F. Russo, D. Veneziano, S. D. Bella, R. Giugno et al., Extracellular circulating viral microRNAs: current knowledge and perspectives, Front Genet, vol.4, p.120, 2013.

N. A. Ahsan, G. C. Sampey, B. Lepene, Y. Akpamagbo, R. A. Barclay et al., Presence of Viral RNA and Proteins in Exosomes from Cellular Clones Resistant to Rift Valley Fever Virus Infection, Front Microbiol, vol.7, p.139, 2016.

, Le système immunitaire inné est capable de faire la différence entre les molécules d'ARN du sois et du non-soi grâce à la présence ou non de modifications spécifiques de l'ARN, de restrictions spatiales et séquences spécifiques des ARN. En ce qui concerne la 2'O-méthylation, des études in vitro ont montré que leur incorporation dans des transcrits d'ARN, des siRNA ou des ARNr 18S empêchait l'activation du TLR (107,108)

, ARNt 2'-O-méthylé est un antagoniste de TLR7 (109) avec un profil de reconnaissance de 2

, De plus, il a été démontré que la 2'-O-méthylation agit comme un inhibiteur du TLR7

, Cependant, d'autres espèces bactériennes dépourvues de Gm18, telles que les souches à Gram positif (par exemple, S. aureus) inhibent également la réponse à TLR7. Cela montre que les, vol.2

, O-méthylations à d'autres positions dans l'ARNt peuvent également avoir des propriétés d'immunosuppression (71)

, Ainsi, dans la perspective de ce travail, nous avons entamé une collaboration avec le Pr

, En effet, notre hypothèse est que nos conditions de stress imitent l'environnement pendant l'invasion de l'hôte (conditions de famine ou d'antibiotiques). L'augmentation de Gm18 pourrait être un moyen pour les bactéries de mieux se cacher de leur hôte lors de leur invasion et de mieux l'envahir. Nous avons envoyé des échantillons d'ARNt à partir de conditions contrôle et de stress (chloramphénicol), Nous nous demandions si l'augmentation de Gm18 dans la réponse au stress pouvait induire une meilleure évasion du système inné

, Grâce à la collaboration avec le Pr. A. Dalpke, nous avons la possibilité de travailler avec des souches provenant de patients. Nous souhaitons effectuer la stratégie expérimentale de stress (contrôle, manque de nutriments et conditions aux antibiotiques) sur ces souches afin de savoir si nous sommes en mesure d'obtenir le même comportement sur des bactéries déjà impliquées dans l'invasion d'un hôte. Ils disposent de souches sans aucune résistance mais également de souches résistantes à la gentamycine. Ici, nous aimerions utiliser ces souches en induisant un stress à la gentamycine afin de voir si elles, De plus, nous travaillons sur la souche DH5? couramment utilisée dans les laboratoires de recherche fondamentale

, En résumé de ce chapitre II, les 2'O-méthylations des ARNt de E. coli sont impliquées dans la réponse au stress et pourraient jouer un rôle dans les MoTT ou dans le domaine du système immunitaire inné. Cela crée de nombreuses nouvelles possibilités pour les 2'O-méthylations et

J. D. Watson and F. H. Crick, Molecular structure of nucleic acids. A structure for deoxyribose nucleic acid, Rev Investig Clin Organo Hosp Enfermedades Nutr. avr, vol.55, issue.2, pp.108-117, 1953.

R. Dahm, Discovering DNA: Friedrich Miescher and the early years of nucleic acid research, Hum Genet. janv, vol.122, issue.6, pp.565-81, 2008.

A. Turchinovich, L. Weiz, A. Langheinz, and B. Burwinkel, Characterization of extracellular circulating microRNA, Nucleic Acids Res. 1 sept, vol.39, issue.16, pp.7223-7256, 2011.

K. C. Vickers, B. T. Palmisano, B. M. Shoucri, R. D. Shamburek, and A. T. Remaley, MicroRNAs are Transported in Plasma and Delivered to Recipient Cells by High-Density Lipoproteins, Nat Cell Biol. avr, vol.13, issue.4, pp.423-456, 2011.

B. György, T. G. Szabó, M. Pásztói, Z. Pál, P. Misják et al., Membrane vesicles, current state-of-the-art: emerging role of extracellular vesicles, Cell Mol Life Sci. août, vol.68, issue.16, pp.2667-88, 2011.

F. Momen-heravi, S. J. Getting, and S. A. Moschos, Extracellular vesicles and their nucleic acids for biomarker discovery, Pharmacol Ther. déc, vol.192, pp.170-87, 2018.

D. W. Greening and R. J. Simpson, Understanding extracellular vesicle diversity -current status, Expert Rev Proteomics. nov, vol.15, issue.11, pp.887-910, 2018.

M. P. Zaborowski, L. Balaj, X. O. Breakefield, and C. P. Lai, Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study, Bioscience. 1 août, vol.65, issue.8, pp.783-97, 2015.

R. Crescitelli, C. Lässer, T. G. Szabó, A. Kittel, M. Eldh et al., Distinct RNA profiles in subpopulations of extracellular vesicles: apoptotic bodies, microvesicles and exosomes, J Extracell Vesicles, vol.2, 2013.

A. Saraste and K. Pulkki, Morphologic and biochemical hallmarks of apoptosis, Cardiovasc Res. févr, vol.45, issue.3, pp.528-565, 2000.

M. Hristov, W. Erl, S. Linder, and P. C. Weber, Apoptotic bodies from endothelial cells enhance the number and initiate the differentiation of human endothelial progenitor cells in vitro, Blood, vol.104, issue.9, pp.2761-2767, 2004.

K. Al-nedawi, B. Meehan, and J. Rak, Microvesicles: messengers and mediators of tumor progression, Cell Cycle Georget Tex. 1 juill, vol.8, issue.13, pp.2014-2022, 2009.

P. A. Holme, N. O. Solum, F. Brosstad, M. Røger, and M. Abdelnoor, Demonstration of plateletderived microvesicles in blood from patients with activated coagulation and fibrinolysis using a filtration technique and western blotting, Thromb Haemost, vol.72, issue.5, pp.666-71, 1994.

G. Van-niel, D. Angelo, G. Raposo, and G. , Shedding light on the cell biology of extracellular vesicles, Nat Rev Mol Cell Biol. avr, vol.19, issue.4, pp.213-241, 2018.

M. Colombo, G. Raposo, and C. Théry, Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles, Annu Rev Cell Dev Biol, vol.30, pp.255-89, 2014.

L. Blanc and M. Vidal, New insights into the function of Rab GTPases in the context of exosomal secretion, Small GTPases, vol.9, issue.1-2, pp.95-106, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02307940

E. Cocucci, G. Racchetti, and J. Meldolesi, Shedding microvesicles: artefacts no more, Trends Cell Biol. févr, vol.19, issue.2, pp.43-51, 2009.

E. Turola, R. Furlan, F. Bianco, M. Matteoli, and C. Verderio, Microglial microvesicle secretion and intercellular signaling, Front Physiol, vol.3, p.149, 2012.

C. D'souza-schorey and J. W. Clancy, Tumor-derived microvesicles: shedding light on novel microenvironment modulators and prospective cancer biomarkers, Genes Dev. 15 juin, vol.26, issue.12, pp.1287-99, 2012.

H. T. Mcmahon and E. Boucrot, Membrane curvature at a glance, J Cell Sci. 15 mars, vol.128, issue.6, pp.1065-70, 2015.

M. M. Kozlov, F. Campelo, N. Liska, L. V. Chernomordik, S. J. Marrink et al., Mechanisms shaping cell membranes, Curr Opin Cell Biol. août, vol.29, pp.53-60, 2014.

G. Raposo and W. Stoorvogel, Extracellular vesicles: exosomes, microvesicles, and friends, J Cell Biol. 18 févr, vol.200, issue.4, pp.373-83, 2013.

N. P. Hessvik and A. Llorente, Current knowledge on exosome biogenesis and release, Cell Mol Life Sci CMLS, vol.75, issue.2, pp.193-208, 2018.

A. V. Savelyeva, E. V. Kuligina, D. N. Bariakin, V. V. Kozlov, E. I. Ryabchikova et al., Variety of RNAs in Peripheral Blood Cells, Plasma, and Plasma Fractions, 2017.

R. C. Rennert, F. H. Hochberg, and B. S. Carter, ExRNA in Biofluids as Biomarkers for Brain Tumors, Cell Mol Neurobiol. avr, vol.36, issue.3, pp.353-60, 2016.

V. Muralidharan-chari, J. W. Clancy, A. Sedgwick, D. Souza-schorey, and C. , Microvesicles: mediators of extracellular communication during cancer progression, J Cell Sci. 15 mai, vol.123, pp.1603-1614, 2010.

D. Toro, J. Herschlik, L. Waldner, C. Mongini, and C. , Emerging roles of exosomes in normal and pathological conditions: new insights for diagnosis and therapeutic applications, Front Immunol, vol.6, p.203, 2015.

J. C. Akers, D. Gonda, R. Kim, B. S. Carter, and C. C. Chen, Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies, J Neurooncol. mai, vol.113, issue.1, pp.1-11, 2013.

A. Beach, H. Zhang, M. Z. Ratajczak, and S. S. Kakar, Exosomes: an overview of biogenesis, composition and role in ovarian cancer, J Ovarian Res. 25 janv, vol.7, p.14, 2014.

R. Wubbolts, R. S. Leckie, P. Veenhuizen, G. Schwarzmann, W. Möbius et al., Proteomic and biochemical analyses of human B cell-derived exosomes. Potential implications for their function and multivesicular body formation, J Biol Chem. 28 mars, vol.278, issue.13, pp.10963-72, 2003.

K. Trajkovic, C. Hsu, S. Chiantia, L. Rajendran, D. Wenzel et al., Ceramide triggers budding of exosome vesicles into multivesicular endosomes, Science. 29 févr, vol.319, issue.5867, pp.1244-1251, 2008.

J. F. Brouwers, M. Aalberts, J. Jansen, G. Van-niel, M. H. Wauben et al., Distinct lipid compositions of two types of human prostasomes, Proteomics. mai, vol.13, pp.1660-1666, 2013.

A. S. Balkom-bwm-van,-eisele, D. M. Pegtel, S. Bervoets, and M. C. Verhaar, Quantitative and qualitative analysis of small RNAs in human endothelial cells and exosomes provides insights into localized RNA processing, degradation and sorting, J Extracell Vesicles, vol.29, issue.2015

E. Willms, H. J. Johansson, I. Mäger, Y. Lee, K. Blomberg et al., Cells release subpopulations of exosomes with distinct molecular and biological properties, Sci Rep. 2 mars, vol.6, p.22519, 2016.

C. Gutiérrez-vázquez, C. Villarroya-beltri, M. Mittelbrunn, and F. Sánchez-madrid, Transfer of extracellular vesicles during immune cell-cell interactions, Immunol Rev. janv, vol.251, issue.1, pp.125-167, 2013.

H. Valadi, K. Ekström, A. Bossios, M. Sjöstrand, J. J. Lee et al., Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells, Nat Cell Biol. juin, vol.9, issue.6, pp.654-663, 2007.

A. Yeri, A. Courtright, R. Reiman, E. Carlson, T. Beecroft et al., Total Extracellular Small RNA Profiles from Plasma, Saliva, and Urine of Healthy Subjects, Sci Rep, vol.17, p.44061, 2017.

T. Yuan, X. Huang, M. Woodcock, M. Du, R. Dittmar et al., Plasma extracellular RNA profiles in healthy and cancer patients, Sci Rep. 20 janv, vol.6, 2016.

M. Li, E. Zeringer, T. Barta, J. Schageman, A. Cheng et al., Analysis of the RNA content of the exosomes derived from blood serum and urine and its potential as biomarkers, Disponible sur, vol.26, p.369, 1652.

Y. Guo, K. Vickers, Y. Xiong, S. Zhao, Q. Sheng et al., Comprehensive evaluation of extracellular small RNA isolation methods from serum in high throughput sequencing, BMC Genomics, vol.18

J. E. Freedman, M. Gerstein, M. E. Rozowsky, J. Levy, D. Kitchen et al., Diverse human extracellular RNAs are widely detected in human plasma, Nat Commun, vol.7, p.11106, 2016.

K. Max, K. Bertram, K. M. Akat, K. A. Bogardus, J. Li et al., Human plasma and serum extracellular small RNA reference profiles and their clinical utility, Proc Natl Acad Sci U S A. 5 juin, vol.115, issue.23, pp.5334-5377, 2018.

X. Huang, T. Yuan, M. Tschannen, Z. Sun, H. Jacob et al., Characterization of human plasma-derived exosomal RNAs by deep sequencing, BMC Genomics, vol.14, p.319, 2013.

M. G. Amorim, R. Valieris, R. D. Drummond, M. P. Pizzi, V. M. Freitas et al., A total transcriptome profiling method for plasma-derived extracellular vesicles: applications for liquid biopsies, vol.7, 2017.

S. U. Umu, H. Langseth, C. Bucher-johannessen, B. Fromm, A. Keller et al., A comprehensive profile of circulating RNAs in human serum, RNA Biol, vol.15, issue.2, pp.242-50, 2018.

K. M. Danielson, R. Rubio, F. Abderazzaq, S. Das, and Y. E. Wang, High Throughput Sequencing of Extracellular RNA from Human Plasma, PloS One, vol.12, issue.1, p.164644, 2017.

H. H. Cheng, H. S. Yi, Y. Kim, E. M. Kroh, J. W. Chien et al., Plasma processing conditions substantially influence circulating microRNA biomarker levels, PloS One, vol.8, issue.6, p.64795, 2013.

J. P. Tosar, A. Cayota, E. Eitan, M. K. Halushka, and K. W. Witwer, Ribonucleic artefacts: are some extracellular RNA discoveries driven by cell culture medium components?, J Extracell Vesicles, vol.6, issue.1, p.1272832, 2017.

A. M. Ainsztein, P. J. Brooks, V. G. Dugan, A. Ganguly, M. Guo et al., The NIH Extracellular RNA Communication Consortium, J Extracell Vesicles, vol.4, p.27493, 2015.

A. Galvanin, G. Dostert, L. Ayadi, V. Marchand, É. Velot et al., Diversity and heterogeneity of extracellular RNA in human plasma, Biochimie. 17 mai, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02140106

A. J. Mitchell, W. D. Gray, S. S. Hayek, Y. Ko, S. Thomas et al., Platelets confound the measurement of extracellular miRNA in archived plasma, Sci Rep, vol.13, p.32651, 2016.

B. Mateescu, E. Kowal, B. Balkom, . Van, S. Bartel et al., Obstacles and opportunities in the functional analysis of extracellular vesicle RNAan ISEV position paper, J Extracell Vesicles. 1 janv, vol.6, issue.1, p.1286095, 2017.

J. D. Arroyo, J. R. Chevillet, E. M. Kroh, I. K. Ruf, C. C. Pritchard et al., Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma, Proc Natl Acad Sci, vol.108, issue.12, pp.5003-5011, 2011.

H. M. Colhoun, J. D. Otvos, M. B. Rubens, M. R. Taskinen, S. R. Underwood et al., Lipoprotein subclasses and particle sizes and their relationship with coronary artery calcification in men and women with and without type 1 diabetes, Diabetes. juin, vol.51, issue.6, pp.1949-56, 2002.

F. Royo, K. Schlangen, L. Palomo, E. Gonzalez, J. Conde-vancells et al., Transcriptome of extracellular vesicles released by hepatocytes, PloS One, vol.8, issue.7, p.68693, 2013.

S. Lucas, F. A. Allenson, K. Bernard, V. Castillo, J. Kim et al., Minimally invasive genomic and transcriptomic profiling of visceral cancers by next-generation sequencing of circulating exosomes, Ann Oncol Off J Eur Soc Med Oncol. avr, vol.27, issue.4, pp.635-676, 2016.

D. Khare, N. Goldschmidt, A. Bardugo, D. Gur-wahnon, I. Z. Ben-dov et al., Plasma microRNA profiling: Exploring better biomarkers for lymphoma surveillance, Disponible sur, vol.12, 2017.

J. Van-deun, P. Mestdagh, R. Sormunen, V. Cocquyt, K. Vermaelen et al., The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling, J Extracell Vesicles, vol.18, issue.2014

Y. Tang, Y. Huang, L. Zheng, S. Qin, X. Xu et al., Comparison of isolation methods of exosomes and exosomal RNA from cell culture medium and serum, Int J Mol Med. sept, vol.40, issue.3, pp.834-878, 2017.

M. Ding, C. Wang, X. Lu, C. Zhang, Z. Zhou et al., Comparison of commercial exosome isolation kits for circulating exosomal microRNA profiling, Anal Bioanal Chem. juin, vol.410, issue.16, pp.3805-3819, 2018.

I. Helwa, J. Cai, M. D. Drewry, A. Zimmerman, M. B. Dinkins et al., A Comparative Study of Serum Exosome Isolation Using Differential Ultracentrifugation and Three Commercial Reagents, Disponible sur, vol.12, 2017.

J. D. Arroyo, J. R. Chevillet, E. M. Kroh, I. K. Ruf, C. C. Pritchard et al., Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma, Proc Natl Acad Sci, vol.108, issue.12, pp.5003-5011, 2011.

M. P. Kowalski and T. Krude, Functional roles of non-coding Y RNAs, Int J Biochem Cell Biol. sept, vol.66, pp.20-29, 2015.

J. M. Dhahbi, S. R. Spindler, H. Atamna, D. Boffelli, P. Mote et al., 5?-YRNA fragments derived by processing of transcripts from specific YRNA genes and pseudogenes are abundant in human serum and plasma, Physiol Genomics, vol.45, issue.21, pp.990-998, 2013.

C. P. Christov, E. Trivier, and T. Krude, Noncoding human Y RNAs are overexpressed in tumours and required for cell proliferation, Br J Cancer. 11 mars, vol.98, issue.5, pp.981-989, 2008.

Y. Motorin and M. Helm, tRNA stabilization by modified nucleotides, Biochemistry. 22 juin, vol.49, issue.24, pp.4934-4978, 2010.

B. El-yacoubi, M. Bailly, and V. De-crécy-lagard, Biosynthesis and function of posttranscriptional modifications of transfer RNAs, Annu Rev Genet, vol.46, pp.69-95, 2012.

H. Grosjean and . Dna, RNA Modification Enzymes : Structure, Mechanism, Function and Evolution, 2009.

G. R. Björk, J. U. Ericson, C. E. Gustafsson, T. G. Hagervall, Y. H. Jönsson et al., Transfer RNA modification, Annu Rev Biochem, vol.56, pp.263-87, 1987.

S. Gehrig, M. Eberle, F. Botschen, K. Rimbach, F. Eberle et al., Identification of modifications in microbial, native tRNA that suppress immunostimulatory activity, J Exp Med. 13 févr, vol.209, issue.2, pp.225-258, 2012.

K. Rimbach, S. Kaiser, M. Helm, A. H. Dalpke, and T. Eigenbrod, 2'-O-Methylation within Bacterial RNA Acts as Suppressor of TLR7/TLR8 Activation in Human Innate Immune Cells, J Innate Immun, vol.7, issue.5, pp.482-93, 2015.

J. Shepherd and M. Ibba, Bacterial transfer RNAs, FEMS Microbiol Rev. mai, vol.39, issue.3, pp.280-300, 2015.

L. Ayadi, A. Galvanin, F. Pichot, V. Marchand, and Y. Motorin, RNA ribose methylation (2'-Omethylation): Occurrence, biosynthesis and biological functions, Biochim Biophys Acta Gene Regul Mech. mars, vol.1862, issue.3, pp.253-69, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01969343

M. A. Machnicka, K. Milanowska, O. Oglou, O. Purta, E. Kurkowska et al., MODOMICS: a database of RNA modification pathways--2013 update, Nucleic Acids Res. janv, vol.41, pp.262-267, 2013.

H. L. Schubert, R. M. Blumenthal, and X. Cheng, Many paths to methyltransfer: a chronicle of convergence, Trends Biochem Sci. juin, vol.28, issue.6, pp.329-364, 2003.

S. Boschi-muller and Y. Motorin, Chemistry enters nucleic acids biology: enzymatic mechanisms of RNA modification, Biochem Biokhimiia. déc, vol.78, issue.13, pp.1392-404, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01467256

P. Z. Kozbial and A. R. Mushegian, Natural history of S-adenosylmethionine-binding proteins, BMC Struct Biol, vol.5, p.19, 2005.

B. C. Persson, G. Jäger, and C. Gustafsson, The spoU gene of Escherichia coli, the fourth gene of the spoT operon, is essential for tRNA (Gm18) 2'-O-methyltransferase activity, Nucleic Acids Res, vol.25, issue.20, pp.4093-4100, 1997.

I. Kumagai, K. Watanabe, and T. Oshima, A thermostable tRNA (guanosine-2')-methyltransferase from Thermus thermophilus HB27 and the effect of ribose methylation on the conformational stability of tRNA, J Biol Chem. 10 juill, vol.257, issue.13, pp.7388-95, 1982.

K. Watanabe, O. Nureki, S. Fukai, R. Ishii, H. Okamoto et al., Roles of conserved amino acid sequence motifs in the SpoU (TrmH) RNA methyltransferase family, J Biol Chem. 18 mars, vol.280, issue.11, pp.10368-77, 2005.

O. Nureki, K. Watanabe, S. Fukai, R. Ishii, Y. Endo et al., Deep knot structure for construction of active site and cofactor binding site of tRNA modification enzyme, Struct Lond Engl, vol.12, issue.4, pp.593-602, 1993.

E. Purta, F. Van-vliet, K. L. Tkaczuk, S. Dunin-horkawicz, H. Mori et al., The yfhQ gene of Escherichia coli encodes a tRNA:Cm32/Um32 methyltransferase, BMC Mol Biol. 18 juill, vol.7, p.23, 2006.

R. Liu, T. Long, M. Zhou, X. Zhou, and E. Wang, tRNA recognition by a bacterial tRNA Xm32 modification enzyme from the SPOUT methyltransferase superfamily, Nucleic Acids Res. 3 sept, vol.43, issue.15, pp.7489-503, 2015.

A. Benítez-páez, M. Villarroya, S. Douthwaite, T. Gabaldón, and M. Armengod, YibK is the 2'-O-methyltransferase TrmL that modifies the wobble nucleotide in Escherichia coli tRNA(Leu) isoacceptors. RNA N Y N, vol.16, pp.2131-2174, 2010.

M. Armengod, I. Moukadiri, S. Prado, R. Ruiz-partida, A. Benítez-páez et al., Enzymology of tRNA modification in the bacterial MnmEG pathway, Biochimie. juill, vol.94, issue.7, pp.1510-1530, 2012.

G. Kawai, H. Ue, M. Yasuda, K. Sakamoto, T. Hashizume et al., Relation between functions and conformational characteristics of modified nucleosides found in tRNAs, Nucleic Acids Symp Ser, issue.25, pp.49-50, 1991.

P. Prusiner, N. Yathindra, and M. Sundaralingam, Effect of ribose O(2')-methylation on the conformation of nucleosides and nucleotides, Biochim Biophys Acta, vol.366, issue.2, pp.115-138, 1974.

B. G. Lane and T. Tamaoki, Studies of the chain termini and alkali-stable dinucleotide sequences in 16 s and 28 s ribosomal RNA from L cells, J Mol Biol. 28 juill, vol.27, issue.2, pp.335-383, 1967.

A. R. Trim and J. E. Parker, Preparation, purification and analyses of thirteen alkali-stable dinucleotides from yeast ribonucleic acid, Biochem J. févr, vol.116, issue.4, pp.589-98, 1970.

L. Endres, P. C. Dedon, and T. J. Begley, Codon-biased translation can be regulated by wobblebase tRNA modification systems during cellular stress responses, RNA Biol. 18 avr, vol.12, issue.6, pp.603-617, 2015.

H. Huang and A. K. Hopper, Multiple Layers of Stress-Induced Regulation in tRNA Biology, Disponible sur, vol.6, 2016.

L. Han, Y. Kon, and E. M. Phizicky, Functional importance of ?38 and ?39 in distinct tRNAs, amplified for tRNAGln(UUG) by unexpected temperature sensitivity of the s2U modification in yeast, RNA N Y N. févr, vol.21, issue.2, pp.188-201, 2015.

M. A. Preston, S. Silva, Y. Kon, and E. M. Phizicky, tRNAHis 5-methylcytidine levels increase in response to several growth arrest conditions in Saccharomyces cerevisiae, RNA N Y N. févr, vol.19, issue.2, pp.243-56, 2013.

F. Alings, L. P. Sarin, C. Fufezan, H. Drexler, and S. A. Leidel, An evolutionary approach uncovers a diverse response of tRNA 2-thiolation to elevated temperatures in yeast, RNA N Y N. févr, vol.21, issue.2, pp.202-214, 2015.

J. R. Damon, D. Pincus, and H. L. Ploegh, tRNA thiolation links translation to stress responses in Saccharomyces cerevisiae, Mol Biol Cell. 15 janv, vol.26, issue.2, pp.270-82, 2015.

K. R. Noon, R. Guymon, P. F. Crain, J. A. Mccloskey, M. Thomm et al., Influence of Temperature on tRNA Modification in Archaea: Methanococcoides burtonii (Optimum Growth Temperature [Topt], 23°C) and Stetteria hydrogenophila (Topt, 95°C), J Bacteriol. sept, vol.185, issue.18, pp.5483-90, 2003.

V. Emilsson, A. K. Näslund, and C. G. Kurland, Thiolation of transfer RNA in Escherichia coli varies with growth rate, Nucleic Acids Res. 11 sept, vol.20, issue.17, pp.4499-505, 1992.

J. Lu, A. Esberg, B. Huang, and A. S. Byström, Kluyveromyces lactis gamma-toxin, a ribonuclease that recognizes the anticodon stem loop of tRNA, Nucleic Acids Res. mars, vol.36, issue.4, pp.1072-80, 2008.

M. Schaefer, T. Pollex, K. Hanna, F. Tuorto, M. Meusburger et al., RNA methylation by Dnmt2 protects transfer RNAs against stress-induced cleavage, Genes Dev. 1 août, vol.24, issue.15, pp.1590-1595, 2010.

A. Alexandrov, I. Chernyakov, W. Gu, S. L. Hiley, T. R. Hughes et al., Rapid tRNA decay can result from lack of nonessential modifications, Mol Cell. 6 janv, vol.21, issue.1, pp.87-96, 2006.

A. Alexandrov, M. R. Martzen, and E. M. Phizicky, Two proteins that form a complex are required for 7-methylguanosine modification of yeast tRNA. RNA N Y N, vol.8, pp.1253-66, 2002.

A. Alexandrov, E. J. Grayhack, and E. M. Phizicky, tRNA m7G methyltransferase Trm8p/Trm82p: evidence linking activity to a growth phenotype and implicating Trm82p in maintaining levels of active Trm8p, RNA N Y N. mai, vol.11, issue.5, pp.821-851, 2005.

C. Chan, M. Dyavaiah, M. S. Demott, K. Taghizadeh, P. C. Dedon et al., A quantitative systems approach reveals dynamic control of tRNA modifications during cellular stress, PLoS Genet. 16 déc, vol.6, issue.12, p.1001247, 2010.

C. Gu, T. J. Begley, and P. C. Dedon, tRNA modifications regulate translation during cellular stress, FEBS Lett, vol.588, issue.23, pp.4287-96, 2014.

V. Marchand, F. Pichot, K. Thüring, L. Ayadi, I. Freund et al., Next-Generation Sequencing-Based RiboMethSeq Protocol for Analysis of tRNA 2?-O-Methylation
URL : https://hal.archives-ouvertes.fr/hal-01799272

. Biomolecules, Disponible sur, vol.7, 2017.

A. Galvanin, L. Ayadi, M. Helm, Y. Motorin, and V. Marchand, Mapping and Quantification of tRNA 2'-O-Methylation by RiboMethSeq, Methods Mol Biol Clifton NJ, vol.1870, pp.273-95, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01957102

S. Jung, T. Von-thülen, V. Laukemper, S. Pigisch, D. Hangel et al., A single naturally occurring 2'-O-methylation converts a TLR7-and TLR8-activating RNA into a TLR8-specific ligand, PloS One, vol.10, issue.3, p.120498, 2015.

A. D. Judge, G. Bola, A. Lee, and I. Maclachlan, Design of noninflammatory synthetic siRNA mediating potent gene silencing in vivo, Mol Ther J Am Soc Gene Ther. mars, vol.13, issue.3, pp.494-505, 2006.

M. Robbins, A. Judge, L. Liang, K. Mcclintock, E. Yaworski et al., 2'-O-methylmodified RNAs act as TLR7 antagonists, Mol Ther J Am Soc Gene Ther. sept, vol.15, issue.9, pp.1663-1672, 2007.

S. Kaiser, K. Rimbach, T. Eigenbrod, A. H. Dalpke, and M. Helm, A modified dinucleotide motif specifies tRNA recognition by TLR7, RNA. sept, vol.20, issue.9, pp.1351-1356, 2014.