M. Eigen, Viral Quasispecies, Scientific American, vol.269, issue.1, pp.42-49, 1993.
DOI : 10.1038/scientificamerican0793-42

J. Drake and J. Holland, Mutation rates among RNA viruses, Proceedings of the National Academy of Sciences, vol.96, issue.24, pp.13910-13913, 1999.
DOI : 10.1073/pnas.96.24.13910

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC24164

J. Holland, D. L. Torre, J. Steinhauer, and D. , RNA Virus Populations as Quasispecies, Curr Top Microbiol Immunol, vol.176, pp.1-20, 1992.
DOI : 10.1007/978-3-642-77011-1_1

A. Lauring and R. Andino, Quasispecies Theory and the Behavior of RNA Viruses, PLoS Pathogens, vol.7, issue.7, p.1001005, 2010.
DOI : 10.1371/journal.ppat.1001005.g004

J. Pfeiffer and K. Kirkegaard, A single mutation in poliovirus RNA-dependent RNA polymerase confers resistance to mutagenic nucleotide analogs via increased fidelity, Proceedings of the National Academy of Sciences, vol.100, issue.12, pp.7289-7294, 2003.
DOI : 10.1073/pnas.1232294100

N. Gnadig, S. Beaucourt, G. Campagnola, A. Borderia, and M. Sanz-ramos, Coxsackievirus B3 mutator strains are attenuated in vivo, Proceedings of the National Academy of Sciences, vol.109, issue.34, pp.2294-2303, 2012.
DOI : 10.1073/pnas.1204022109

URL : https://hal.archives-ouvertes.fr/pasteur-00918212

R. Graham, M. Becker, L. Eckerle, M. Bolles, and M. Denison, A live, impaired-fidelity coronavirus vaccine protects in an aged, immunocompromised mouse model of lethal disease, Nature Medicine, vol.110, issue.12, pp.1820-1826, 2012.
DOI : 10.1073/pnas.0808116105

J. Arnold, M. Vignuzzi, J. Stone, R. Andino, and C. Cameron, Remote Site Control of an Active Site Fidelity Checkpoint in a Viral RNA-dependent RNA Polymerase, Journal of Biological Chemistry, vol.280, issue.27, pp.25706-25716, 2005.
DOI : 10.1074/jbc.M503444200

C. Ferrer-orta, M. Sierra, R. Agudo, I. De-la-higuera, and A. Arias, Structure of Foot-and-Mouth Disease Virus Mutant Polymerases with Reduced Sensitivity to Ribavirin, Journal of Virology, vol.84, issue.12, pp.6188-6199, 2010.
DOI : 10.1128/JVI.02420-09

L. Mansky and K. Cunningham, Virus mutators and antimutators, Trends in Genetics, vol.16, issue.11, pp.512-517, 2000.
DOI : 10.1016/S0168-9525(00)02125-9

J. Speyer, Mutagenic DNA polymerase, Biochemical and Biophysical Research Communications, vol.21, issue.1, pp.6-8, 1965.
DOI : 10.1016/0006-291X(65)90417-1

J. Drake, E. Allen, S. Forsberg, R. Preparata, and E. Greening, Spontaneous Mutation: Genetic Control of Mutation Rates in Bacteriophage T4, Nature, vol.55, issue.5186, pp.1128-1132, 1969.
DOI : 10.1016/0042-6822(68)90275-4

P. Suarez, J. Valcarcel, and J. Ortin, Heterogeneity of the mutation rates of influenza A viruses: isolation of mutator mutants, J Virol, vol.66, pp.2491-2494, 1992.

S. Sadeghipour, E. Bek, and P. Mcminn, Ribavirin-Resistant Mutants of Human Enterovirus 71 Express a High Replication Fidelity Phenotype during Growth in Cell Culture, Journal of Virology, vol.87, issue.3, pp.1759-1769, 2013.
DOI : 10.1128/JVI.02139-12

V. Furio, A. Moya, and R. Sanjuan, The cost of replication fidelity in human immunodeficiency virus type 1, Proceedings of the Royal Society B: Biological Sciences, vol.274, issue.4, pp.225-230, 2007.
DOI : 10.1016/S0092-8674(00)81191-5

V. Furio, A. Moya, and R. Sanjuan, The cost of replication fidelity in an RNA virus, Proceedings of the National Academy of Sciences, vol.102, issue.29, pp.10233-10237, 2005.
DOI : 10.1073/pnas.0501062102

S. Weeks, C. Lee, Y. Zhao, E. Smidansky, and A. August, A Polymerase Mechanism-based Strategy for Viral Attenuation and Vaccine Development, Journal of Biological Chemistry, vol.287, issue.38, pp.31618-31622, 2012.
DOI : 10.1074/jbc.C112.401471

P. Gong and O. Peersen, Structural basis for active site closure by the poliovirus RNA-dependent RNA polymerase, Proceedings of the National Academy of Sciences, vol.107, issue.52, pp.22505-22510, 2010.
DOI : 10.1073/pnas.1007626107

M. Vignuzzi, J. Stone, J. Arnold, C. Cameron, and R. Andino, Quasispecies diversity determines pathogenesis through cooperative interactions in a viral population, Nature, vol.77, issue.7074, pp.344-348, 2006.
DOI : 10.1038/nature04388

E. Holmes, Error thresholds and the constraints to RNA virus evolution, Trends in Microbiology, vol.11, issue.12, pp.543-546, 2003.
DOI : 10.1016/j.tim.2003.10.006

H. Edajj, Mutation rates and rapid evolution of RNA viruses The Evolutionary Biology of Viruses, pp.161-184, 1994.

J. Holland, E. Domingo, J. De-la-torre, and D. Steinhauer, Mutation frequencies at defined single codon sites in vesicular stomatitis virus and poliovirus can be increased only slightly by chemical mutagenesis, J Virol, vol.64, pp.3960-3962, 1990.

L. Loeb, J. Essigmann, F. Kazazi, J. Zhang, and K. Rose, Lethal mutagenesis of HIV with mutagenic nucleoside analogs, Proceedings of the National Academy of Sciences, vol.96, issue.4, pp.1492-1497, 1999.
DOI : 10.1073/pnas.96.4.1492

J. Pfeiffer and K. Kirkegaard, Increased Fidelity Reduces Poliovirus Fitness and Virulence under Selective Pressure in Mice, PLoS Pathogens, vol.78, issue.2, p.11, 2005.
DOI : 0027-8424(1981)078[4887:MCOPCA]2.0.CO;2

L. Coffey, Y. Beeharry, A. Borderia, H. Blanc, and M. Vignuzzi, Arbovirus high fidelity variant loses fitness in mosquitoes and mice, Proceedings of the National Academy of Sciences, vol.108, issue.38, pp.16038-16043, 2011.
DOI : 10.1073/pnas.1111650108

URL : https://hal.archives-ouvertes.fr/pasteur-00620624

U. Schultz, W. Fitch, S. Ludwig, J. Mandler, and C. Scholtissek, Evolution of pig influenza viruses, Virology, vol.183, issue.1, pp.61-73, 1991.
DOI : 10.1016/0042-6822(91)90118-U

M. Vignuzzi, J. Stone, and R. Andino, Ribavirin and lethal mutagenesis of poliovirus: molecular mechanisms, resistance and biological implications, Virus Research, vol.107, issue.2, pp.173-181, 2005.
DOI : 10.1016/j.virusres.2004.11.007

J. Anderson, R. Daifuku, and L. Loeb, Viral Error Catastrophe by Mutagenic Nucleosides, Annual Review of Microbiology, vol.58, issue.1, pp.183-205, 2004.
DOI : 10.1146/annurev.micro.58.030603.123649

J. Bull, R. Sanjuan, and C. Wilke, Theory of Lethal Mutagenesis for Viruses, Journal of Virology, vol.81, issue.6, pp.2930-2939, 2007.
DOI : 10.1128/JVI.01624-06

J. Graci and C. Cameron, Therapeutically targeting RNA viruses via lethal mutagenesis, Future Virology, vol.3, issue.6, pp.553-566, 2008.
DOI : 10.2217/17460794.3.6.553

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2630198

M. Dapp, S. Patterson, and L. Mansky, Back to the future: revisiting HIV-1 lethal mutagenesis, Trends in Microbiology, vol.21, issue.2, pp.56-62, 2012.
DOI : 10.1016/j.tim.2012.10.006

M. Dapp, R. Heineman, and L. Mansky, Interrelationship between HIV-1 Fitness and Mutation Rate, Journal of Molecular Biology, vol.425, issue.1, pp.41-53, 2013.
DOI : 10.1016/j.jmb.2012.10.009

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3534800

L. Coffey and M. Vignuzzi, Host Alternation of Chikungunya Virus Increases Fitness while Restricting Population Diversity and Adaptability to Novel Selective Pressures, Journal of Virology, vol.85, issue.2, pp.1025-1035, 2011.
DOI : 10.1128/JVI.01918-10

URL : https://hal.archives-ouvertes.fr/pasteur-00546712

L. Coffey, N. Forrester, K. Tsetsarkin, N. Vasilakis, and S. Weaver, Factors shaping the adaptive landscape for arboviruses: implications for the emergence of disease, Future Microbiology, vol.8, issue.2, pp.155-176, 2013.
DOI : 10.2217/fmb.12.139

L. Coffey, N. Vasilakis, A. Brault, A. Powers, and F. Tripet, Arbovirus evolution in vivo is constrained by host alternation, Proceedings of the National Academy of Sciences, vol.105, issue.19, pp.6970-6975, 2008.
DOI : 10.1073/pnas.0712130105

A. Ciota, A. Lovelace, Y. Jia, L. Davis, and D. Young, Characterization of mosquito-adapted West Nile virus, Journal of General Virology, vol.89, issue.7, pp.1633-1642, 2008.
DOI : 10.1099/vir.0.2008/000893-0

I. Greene, E. Wang, E. Deardorff, R. Milleron, and E. Domingo, Effect of Alternating Passage on Adaptation of Sindbis Virus to Vertebrate and Invertebrate Cells, Journal of Virology, vol.79, issue.22, pp.14253-14260, 2005.
DOI : 10.1128/JVI.79.22.14253-14260.2005

E. Deardorff, K. Fitzpatrick, G. Jerzak, P. Shi, and L. Kramer, West Nile Virus Experimental Evolution in vivo and the Trade-off Hypothesis, PLoS Pathogens, vol.146, issue.11, p.1002335, 2011.
DOI : 10.1371/journal.ppat.1002335.t001

N. Forrester, M. Guerbois, R. Seymour, H. Spratt, and S. Weaver, Vector-Borne Transmission Imposes a Severe Bottleneck on an RNA Virus Population, PLoS Pathogens, vol.8, issue.9, p.1002897, 2012.
DOI : 10.1371/journal.ppat.1002897.s007

S. Weaver, R. Rico-hesse, and T. Scott, Genetic Diversity and Slow Rates of Evolution in New World Alphaviruses, Curr Top Microbiol Immunol, vol.176, pp.99-117, 1992.
DOI : 10.1007/978-3-642-77011-1_7

A. Ciota, K. Ngo, A. Lovelace, A. Payne, and Y. Zhou, Role of the mutant spectrum in adaptation and replication of West Nile virus, Journal of General Virology, vol.88, issue.3, pp.865-874, 2007.
DOI : 10.1099/vir.0.82606-0

S. Lin, S. Hsieh, Y. Yueh, T. Lin, and D. Chao, Study of Sequence Variation of Dengue Type 3 Virus in Naturally Infected Mosquitoes and Human Hosts: Implications for Transmission and Evolution, Journal of Virology, vol.78, issue.22, pp.12717-12721, 2004.
DOI : 10.1128/JVI.78.22.12717-12721.2004

G. Jerzak, K. Bernard, L. Kramer, and G. Ebel, Genetic variation in West Nile virus from naturally infected mosquitoes and birds suggests quasispecies structure and strong purifying selection, Journal of General Virology, vol.86, issue.8, pp.2175-2183, 2005.
DOI : 10.1099/vir.0.81015-0

T. Kurosu, Quasispecies of dengue virus, Tropical Medicine and Health, vol.62, issue.5, pp.29-36, 2011.
DOI : 10.1006/viro.2002.1635

A. Ciota, D. Ehrbar, G. Van-slyke, G. Willsey, and L. Kramer, Cooperative interactions in the West Nile virus mutant swarm, BMC Evolutionary Biology, vol.12, issue.1, p.58, 2012.
DOI : 10.1006/viro.1999.0056

Y. Karpe, P. Aher, and K. Lole, NTPase and 5???-RNA Triphosphatase Activities of Chikungunya Virus nsP2 Protein, PLoS ONE, vol.92, issue.7, p.22336, 2011.
DOI : 10.1371/journal.pone.0022336.t002

A. Rathore, M. Ng, and S. Vasudevan, Differential unfolded protein response during Chikungunya and Sindbis virus infection: CHIKV nsP4 suppresses eIF2?? phosphorylation, Virology Journal, vol.10, issue.1, p.36, 2013.
DOI : 10.1016/0303-2647(89)90053-1

URL : http://doi.org/10.1186/1743-422x-10-36

H. Malet, B. Coutard, S. Jamal, H. Dutartre, and N. Papageorgiou, The Crystal Structures of Chikungunya and Venezuelan Equine Encephalitis Virus nsP3 Macro Domains Define a Conserved Adenosine Binding Pocket, Journal of Virology, vol.83, issue.13, pp.6534-6545, 2009.
DOI : 10.1128/JVI.00189-09

J. Fros, N. Domeradzka, J. Baggen, C. Geertsema, and J. Flipse, Chikungunya Virus nsP3 Blocks Stress Granule Assembly by Recruitment of G3BP into Cytoplasmic Foci, Journal of Virology, vol.86, issue.19, pp.10873-10879, 2012.
DOI : 10.1128/JVI.01506-12

P. Jones, M. Maric, M. Madison, M. W. Roller, and R. , BST-2/tetherin-mediated restriction of chikungunya (CHIKV) VLP budding is counteracted by CHIKV non-structural protein 1 (nsP1), Virology, vol.438, issue.1, pp.37-49, 2013.
DOI : 10.1016/j.virol.2013.01.010

O. Schwartz and M. Albert, Biology and pathogenesis of chikungunya virus, Nature Reviews Microbiology, vol.81, issue.7, pp.491-500, 2010.
DOI : 10.1038/nrmicro2368

URL : https://hal.archives-ouvertes.fr/pasteur-00498486

M. Solignat, B. Gay, S. Higgs, L. Briant, and C. Devaux, Replication cycle of chikungunya: A re-emerging arbovirus, Virology, vol.393, issue.2, pp.183-197, 2009.
DOI : 10.1016/j.virol.2009.07.024

URL : https://hal.archives-ouvertes.fr/hal-00420502

. Mayuri, T. Geders, J. Smith, and R. Kuhn, Role for Conserved Residues of Sindbis Virus Nonstructural Protein 2 Methyltransferase-Like Domain in Regulation of Minus-Strand Synthesis and Development of Cytopathic Infection, Journal of Virology, vol.82, issue.15, pp.7284-7297, 2008.
DOI : 10.1128/JVI.00224-08

J. Lemm and C. Rice, Roles of nonstructural polyproteins and cleavage products in regulating Sindbis virus RNA replication and transcription, J Virol, vol.67, pp.1916-1926, 1993.

K. Saxton-shaw, J. Ledermann, E. Borland, J. Stovall, and E. Mossel, O'nyong nyong Virus Molecular Determinants of Unique Vector Specificity Reside in Non-Structural Protein 3, PLoS Neglected Tropical Diseases, vol.82, issue.1, p.1931, 2013.
DOI : 10.1371/journal.pntd.0001931.s007

D. Kim, A. Firth, S. Atasheva, E. Frolova, and I. Frolov, Conservation of a Packaging Signal and the Viral Genome RNA Packaging Mechanism in Alphavirus Evolution, Journal of Virology, vol.85, issue.16, pp.8022-8036, 2011.
DOI : 10.1128/JVI.00644-11

S. Tomar, R. Hardy, J. Smith, and R. Kuhn, Catalytic Core of Alphavirus Nonstructural Protein nsP4 Possesses Terminal Adenylyltransferase Activity, Journal of Virology, vol.80, issue.20, pp.9962-9969, 2006.
DOI : 10.1128/JVI.01067-06

S. Crotty, D. Maag, J. Arnold, W. Zhong, and J. Lau, The broadspectrum antiviral ribonucleoside ribavirin is an RNA virus mutagen, Nat Med, vol.6, pp.1375-1379, 2000.

C. Cameron and C. Castro, The mechanism of action of ribavirin: lethal mutagenesis of RNA virus genomes mediated by the viral RNA-dependent RNA polymerase, Current Opinion in Infectious Diseases, vol.14, issue.6, pp.757-764, 2001.
DOI : 10.1097/00001432-200112000-00015

K. Mcknight, D. Simpson, S. Lin, T. Knott, and J. Polo, Deduced consensus sequence of Sindbis virus strain AR339: mutations contained in laboratory strains which affect cell culture and in vivo phenotypes, J Virol, vol.70, pp.1981-1989, 1996.

S. Beaucourt, A. Borderia, L. Coffey, N. Gnadig, and M. Sanz-ramos, Isolation of Fidelity Variants of RNA Viruses and Characterization of Virus Mutation Frequency, Journal of Visualized Experiments, issue.52, p.2953, 2011.
DOI : 10.3791/2953

URL : https://hal.archives-ouvertes.fr/pasteur-00601728

E. Aronesty, Command-line tools for processing biological sequencing data Available: http://code.google.com, 2011.

H. Li and R. Durbin, Fast and accurate short read alignment with Burrows-Wheeler transform, Bioinformatics, vol.25, issue.14, pp.1754-1760, 2009.
DOI : 10.1093/bioinformatics/btp324

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2705234

H. Li, B. Handsaker, A. Wysoker, T. Fennell, and J. Ruan, The Sequence Alignment/Map format and SAMtools, Bioinformatics, vol.25, issue.16, pp.2078-2079, 2009.
DOI : 10.1093/bioinformatics/btp352

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2723002

K. Li, E. Venter, S. Yooseph, T. Stockwell, and L. Eckerle, ANDES: Statistical tools for the ANalyses of DEep Sequencing, BMC Research Notes, vol.3, issue.1, p.199, 2010.
DOI : 10.1186/1756-0500-3-199

P. Pal, K. Dowd, J. Brien, M. Edeling, and S. Gorlatov, Development of a Highly Protective Combination Monoclonal Antibody Therapy against Chikungunya Virus, PLoS Pathogens, vol.60, issue.Pt 12, p.1003312, 2013.
DOI : 10.1371/journal.ppat.1003312.s009

R. Lanciotti, O. Kosoy, J. Laven, A. Panella, and J. Velez, Chikungunya Virus in US Travelers Returning from India, 2006, Emerging Infectious Diseases, vol.13, issue.5, pp.764-767, 2006.
DOI : 10.3201/eid1305.070015

A. Powers and C. Logue, Changing patterns of chikungunya virus: re-emergence of a zoonotic arbovirus, Journal of General Virology, vol.88, issue.9, pp.2363-2377, 2007.
DOI : 10.1099/vir.0.82858-0

S. Cherry and N. Perrimon, Entry is a rate-limiting step for viral infection in a Drosophila melanogaster model of pathogenesis, Nature Immunology, vol.5, issue.1, pp.81-87, 2004.
DOI : 10.1038/ni1019

M. Denison, R. Graham, E. Donaldson, L. Eckerle, and R. Baric, Coronaviruses, RNA Biology, vol.70, issue.2, pp.270-279, 2011.
DOI : 10.1128/JVI.79.4.2346-2355.2005

URL : https://hal.archives-ouvertes.fr/pasteur-00918207

M. Vignuzzi, E. Wendt, and R. Andino, Engineering attenuated virus vaccines by controlling replication fidelity, Nature Medicine, vol.7, issue.2, pp.154-161, 2008.
DOI : 10.1038/nm1726

A. Arias, J. Arnold, M. Sierra, E. Smidansky, and E. Domingo, Determinants of RNA-Dependent RNA Polymerase (In)fidelity Revealed by Kinetic Analysis of the Polymerase Encoded by a Foot-and-Mouth Disease Virus Mutant with Reduced Sensitivity to Ribavirin, Journal of Virology, vol.82, issue.24, pp.12346-12355, 2008.
DOI : 10.1128/JVI.01297-08

V. Korneeva and C. Cameron, Structure-Function Relationships of the Viral RNA-dependent RNA Polymerase: FIDELITY, REPLICATION SPEED, AND INITIATION MECHANISM DETERMINED BY A RESIDUE IN THE RIBOSE-BINDING POCKET, Journal of Biological Chemistry, vol.282, issue.22, pp.16135-16145, 2007.
DOI : 10.1074/jbc.M610090200

S. Crotty, C. Cameron, and R. Andino, RNA virus error catastrophe: Direct molecular test by using ribavirin, Proceedings of the National Academy of Sciences, vol.98, issue.12, pp.6895-6900, 2001.
DOI : 10.1073/pnas.111085598

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC34449

N. Vasilakis, E. Deardorff, J. Kenney, S. Rossi, and K. Hanley, Mosquitoes Put the Brake on Arbovirus Evolution: Experimental Evolution Reveals Slower Mutation Accumulation in Mosquito Than Vertebrate Cells, PLoS Pathogens, vol.79, issue.1, p.1000467, 2009.
DOI : 10.1371/journal.ppat.1000467.t011

G. Jerzak, K. Bernard, L. Kramer, P. Shi, and G. Ebel, The West Nile virus mutant spectrum is host-dependant and a determinant of mortality in mice, Virology, vol.360, issue.2, pp.469-476, 2007.
DOI : 10.1016/j.virol.2006.10.029

G. Jerzak, I. Brown, P. Shi, L. Kramer, and G. Ebel, Genetic diversity and purifying selection in West Nile virus populations are maintained during host switching, Virology, vol.374, issue.2, pp.256-260, 2008.
DOI : 10.1016/j.virol.2008.02.032

L. Bertolotti, U. Kitron, E. Walker, M. Ruiz, and J. Brawn, Fine-scale genetic variation and evolution of West Nile Virus in a transmission ???hot spot??? in suburban Chicago, USA, Virology, vol.374, issue.2, pp.381-389, 2008.
DOI : 10.1016/j.virol.2007.12.040

A. Ciota, A. Lovelace, S. Jones, A. Payne, and L. Kramer, Adaptation of two flaviviruses results in differences in genetic heterogeneity and virus adaptability, Journal of General Virology, vol.88, issue.9, pp.2398-2406, 2007.
DOI : 10.1099/vir.0.83061-0

D. Brackney, K. Pesko, I. Brown, E. Deardorff, and J. Kawatachi, West Nile Virus Genetic Diversity is Maintained during Transmission by Culex pipiens quinquefasciatus Mosquitoes, PLoS ONE, vol.2, issue.9, p.24466, 2011.
DOI : 10.1371/journal.pone.0024466.s005

URL : http://doi.org/10.1371/journal.pone.0024466

P. Rose, S. Hanna, A. Spiridigliozzi, N. Wannissorn, and D. Beiting, Natural Resistance-Associated Macrophage Protein Is a Cellular Receptor for Sindbis Virus in Both Insect and Mammalian Hosts, Cell Host & Microbe, vol.10, issue.2, pp.97-104, 2011.
DOI : 10.1016/j.chom.2011.06.009

S. Merkling and R. Van-rij, Beyond RNAi: Antiviral defense strategies in Drosophila and mosquito, Journal of Insect Physiology, vol.59, issue.2, pp.159-170, 2013.
DOI : 10.1016/j.jinsphys.2012.07.004

R. Sanjuan, M. Nebot, N. Chirico, L. Mansky, and R. Belshaw, Viral Mutation Rates, Journal of Virology, vol.84, issue.19, pp.9733-9748, 2010.
DOI : 10.1128/JVI.00694-10

C. Castro, J. Arnold, and C. Cameron, Incorporation fidelity of the viral RNA-dependent RNA polymerase: a kinetic, thermodynamic and structural perspective, Virus Research, vol.107, issue.2, pp.141-149, 2005.
DOI : 10.1016/j.virusres.2004.11.004

J. Arnold, S. Ghosh, and C. Cameron, Poliovirus RNA-dependent RNA Polymerase (3Dpol): DIVALENT CATION MODULATION OF PRIMER, TEMPLATE, AND NUCLEOTIDE SELECTION, Journal of Biological Chemistry, vol.274, issue.52, pp.37060-37069, 1999.
DOI : 10.1074/jbc.274.52.37060

M. Freistadt, J. Vaccaro, and K. Eberle, Biochemical characterization of the fidelity of poliovirus RNA-dependent RNA polymerase, Virology Journal, vol.4, issue.1, p.44, 2007.
DOI : 10.1186/1743-422X-4-44

L. Levi, N. Gnadig, S. Beaucourt, M. Mcpherson, and B. Baron, Fidelity Variants of RNA Dependent RNA Polymerases Uncover an Indirect, Mutagenic Activity of Amiloride Compounds, PLoS Pathogens, vol.104, issue.Pt 1, p.1001163, 2010.
DOI : 10.1371/journal.ppat.1001163.s004

URL : https://hal.archives-ouvertes.fr/pasteur-00530598

W. Klimstra, K. Ryman, and R. Johnston, Adaptation of Sindbis virus to BHK cells selects for use of heparan sulfate as an attachment receptor, J Virol, vol.72, pp.7357-7366, 1998.

K. Bernard, W. Klimstra, and R. Johnston, Mutations in the E2 Glycoprotein of Venezuelan Equine Encephalitis Virus Confer Heparan Sulfate Interaction, Low Morbidity, and Rapid Clearance from Blood of Mice, Virology, vol.276, issue.1, pp.93-103, 2000.
DOI : 10.1006/viro.2000.0546

D. Brackney, J. Beane, and G. Ebel, RNAi Targeting of West Nile Virus in Mosquito Midguts Promotes Virus Diversification, PLoS Pathogens, vol.38, issue.7, p.1000502, 2009.
DOI : 10.1371/journal.ppat.1000502.s004

T. Teo, F. Lum, W. Lee, and L. Ng, Mouse models for Chikungunya virus: deciphering immune mechanisms responsible for disease and pathology, Immunologic Research, vol.19, issue.Suppl 1, pp.136-147, 2012.
DOI : 10.1007/s12026-012-8266-x

A. Ciota, Y. Jia, A. Payne, G. Jerzak, and L. Davis, Experimental Passage of St. Louis Encephalitis Virus In Vivo in Mosquitoes and Chickens Reveals Evolutionarily Significant Virus Characteristics, PLoS ONE, vol.382, issue.1, p.7876, 2009.
DOI : 10.1371/journal.pone.0007876.t003

K. Pesko and G. Ebel, West Nile virus population genetics and evolution, Infection, Genetics and Evolution, vol.12, issue.2, pp.181-190, 2012.
DOI : 10.1016/j.meegid.2011.11.014

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4494105

D. Eisenberg, R. Weiss, T. Terwilliger, and W. Wilcox, Hydrophobic moments and protein structure, Faraday Symposia of the Chemical Society, vol.17, pp.109-120, 1982.
DOI : 10.1039/fs9821700109