#. , S. St, and . Louis, Transfected cells were amplified and subsequently cloned by serial limit dilution At least 5 clones were screened for each RLR to detect the tagged protein expression by qPCR and Western Blot. For STING37shRLR cell lines, we generated lentiviral vectors using the canonical triple transfection of HEK293T cells by a VSVg envelope, an encapsidation p8, G418 at 500 mg

P. Charneau and . Zufferey, MDA5(RHS4430-101128286-V3LHS_300657, shMDA5) or RIG-I (RHS4430-99619609-V2LHS_197176, shRIG-I), all bearing a puromycin selection marker. Vectors were titrated according to manufacturer's instructions in HeLa cells 2013]) was transduced at an MOI of 0.3 and 48 hr later puromycin (5 mg/ml) was added to the media to select properly transduced cells. CHIKV 06?49 strain was isolated from the serum of an adult patient with arthralgia/ myalgia in Saint Louis city, Ré union CHIKV strain 06? 49 was titrated on VERO cell by TCID50 (50% Tissue Culture Infective Dose) Recombinant CHIKV- Rluc which contains the Rluc reporter gene inserted between the non-structural nsP3 and nsP4 proteins has already been described Attenuated MV Schwarz vaccine strain (MV) and recombinant MV Schwarz expressing Fluc (rMV2-Fluc) from an additional transcription unit derived from MV have been previously described To prevent V protein expression from MV, a two-step PCR strategy was used to generate MVDV virus. Two PCR fragments were amplified using MV2313 (5'-ATCTGCTCCCATCTCTATGG) and MV2504 (5'-TCTGTGCCCTTCTTAATGGG) for the first fragment and MV2485 (5'-CCCATTAA- GAAGGGCACAGA) and MV3385 (5'-AGGTTGTACTAGTTGGGTCG) for the second fragment. These PCRs introduced a mutation interfering with RNA editing (UUAAAAAGGGCACAGA native sequence was mutated to UUAAgAAGGGCACAGA) The two PCR products were combined in a second PCR reaction using MV2313 and MV3385 primers. The produced mutated HindIII-SpeI MV fragment was moved into the pTM-MVSchwarz plasmid after digestion with the corresponding restriction enzymes, Thermo Scientific) expressing either an shRNA with no target (RHS4346, shNeg), or targeting LGP2 (shLGP2) and named MV4V. Immunoblot analysis was performed to validate the lack of MV-V expression by MVDV (data not shown). Virus stocks were produced on VERO cells, and titrated by TCID50, 1997.

, Hercules, California) with MOPS running buffer and transferred to cellulose membranes (GE Healthcare, Little Chalfont, United Kingdom) with the Criterion Blotter system (BioRad) The following antibodies were used: an anti-STrEP-Tag (#34850, Qiagen anti-RIG-I (D14G6, Cell Signaling) or monoclonal anti-b-actin antibody (A5441, Sigma), IRF3 (#11904, Cell Signaling) or phosphor-IRF3 (ab76493 HRP-coupled anti-mouse (NA9310V, GE Healthcare) or anti-rabbit (RPN4301, GE Healthcare) were used as secondary antibodies. Peroxidase activity was visualized with an ECL Plus Western Blotting Detection System (#RPN2132, GE Healthcare). MV intracellular staining was performed with mouse anti-N mAb (clone 25 For CHIKV, intracellular staining was performed with either FITC-conjugated anti-CHIK, Antibodies, western blot and intracellular staining Protein extracts were resolved by SDS-polyacrylamide gel electrophoresis on 4?12% Criterion gels (BioRad anti-LGP2 (NBP1- 85348, Novus, Littleton, Colorado), anti-MDA5 (#5321, Cell Signaling, pp.2-1201, 1981.

, 10 5 per well) were plated in a 24-well plate and infected with appropriate MOIs. For rMV2-Fluc or CHIKV-Rluc infection, at each time point cells were lysed by Passive Lysis buffer (E1941, Promega, Fitchburg, Wisconsin), and luciferase activity was measured with Bright- Glow Luciferase assay system (E2650, Promega) or Renilla Glow Luciferase Assay System (E2720, Promega), correspondingly. For immunostaining, cells were washed twice with phosphate-buffered saline (PBS) and 2% foetal calf serum (FCS) and then, MV and CHIKV replication assays STING37shRLR cells

A. Ablasser, F. Bauernfeind, G. Hartmann, E. Latz, K. Fitzgerald et al., RIG-I-dependent sensing of poly(dA:dT) through the induction of an RNA polymerase III???transcribed RNA intermediate, Nature Immunology, vol.97, issue.10, pp.1065-1072, 2009.
DOI : 10.4049/jimmunol.0803001

I. Akhrymuk, S. Kulemzin, and E. Frolova, Evasion of the Innate Immune Response: the Old World Alphavirus nsP2 Protein Induces Rapid Degradation of Rpb1, a Catalytic Subunit of RNA Polymerase II, Journal of Virology, vol.86, issue.13, pp.7180-7191, 2012.
DOI : 10.1128/JVI.00541-12

J. Andrejeva, K. Childs, D. Young, T. Carlos, N. Stock et al., The V proteins of paramyxoviruses bind the IFN-inducible RNA helicase, mda-5, and inhibit its activation of the IFN-?? promoter, Proceedings of the National Academy of Sciences, vol.224, issue.1-2, pp.17264-17269, 2004.
DOI : 10.1016/S0022-1759(99)00017-4

A. Baum, R. Sachidanandam, and A. García-sastre, Preference of RIG-I for short viral RNA molecules in infected cells revealed by next-generation sequencing, Proceedings of the National Academy of Sciences, vol.79, issue.13, pp.16303-16308, 2010.
DOI : 10.1128/JVI.79.13.8431-8439.2005

Y. Benjamini and Y. Hochberg, Controlling the false discovery rate: a practical and powerful approach to multiple testing, Journal of the Royal Statistical Society, vol.57, pp.289-300, 1995.

L. Boura?¨mboura?¨boura?¨m, M. Gad, H. Drosten, C. Jacob, Y. Tafforeau et al., Mapping of Chikungunya virus interactions with host proteins identified nsP2 as a highly connected viral component, Journal of Virology, vol.86, pp.3121-3134, 1128.

A. Bré-hin, L. Rubrecht, M. Navarro-sanchez, V. Maré-chal, M. Frenkiel et al., Production and characterization of mouse monoclonal antibodies reactive to Chikungunya envelope E2 glycoprotein, Virology, vol.371, issue.1, pp.185-195, 2008.
DOI : 10.1016/j.virol.2007.09.028

A. Bruns and C. Horvath, Activation of RIG-I-like receptor signal transduction, Critical Reviews in Biochemistry and Molecular Biology, vol.12, issue.2, pp.194-206, 2011.
DOI : 10.1038/ni.1979

A. Bruns, D. Pollpeter, N. Hadizadeh, S. Myong, J. Marko et al., ATP Hydrolysis Enhances RNA Recognition and Antiviral Signal Transduction by the Innate Immune Sensor, Laboratory of Genetics and Physiology 2 (LGP2), Journal of Biological Chemistry, vol.29, issue.2, pp.938-946, 2013.
DOI : 10.1016/0003-2697(76)90527-3

A. Bruns, G. Leser, R. Lamb, and C. Horvath, The Innate Immune Sensor LGP2 Activates Antiviral Signaling by Regulating MDA5-RNA Interaction and Filament Assembly, Molecular Cell, vol.55, issue.5, pp.771-781, 2014.
DOI : 10.1016/j.molcel.2014.07.003

R. Cattaneo, G. Rebmann, A. Schmid, K. Baczko, V. Ter-meulen et al., Altered transcription of a defective measles virus genome derived from a diseased human brain, EMBO Journal, vol.6, pp.681-688, 1987.

R. Chen, E. Wang, K. Tsetsarkin, and S. Weaver, Chikungunya Virus 3??? Untranslated Region: Adaptation to Mosquitoes and a Population Bottleneck as Major Evolutionary Forces, PLoS Pathogens, vol.6, issue.8, 2013.
DOI : 10.1371/journal.ppat.1003591.s004

G. Cheng, L. Wang, Z. Fridlender, G. Cheng, B. Chen et al., Pharmacologic Activation of the Innate Immune System to Prevent Respiratory Viral Infections, American Journal of Respiratory Cell and Molecular Biology, vol.11, issue.3, pp.480-4882010, 2011.
DOI : 10.1593/neo.09506

K. Childs, R. Randall, and S. Goodbourn, LGP2 plays a critical role in sensitizing mda-5 to activation by doublestranded RNA, PloS One, vol.8, 2013.

Y. Chiu, J. Macmillan, and Z. Chen, RNA Polymerase III Detects Cytosolic DNA and Induces Type I Interferons through the RIG-I Pathway, Cell, vol.138, issue.3, pp.576-591, 2009.
DOI : 10.1016/j.cell.2009.06.015

M. Choi, Z. Wang, T. Ban, H. Yanai, Y. Lu et al., A selective contribution of the RIG-I-like receptor pathway to type I interferon responses activated by cytosolic DNA, Proceedings of the National Academy of Sciences, vol.138, issue.3, pp.17870-17875, 2009.
DOI : 10.1016/j.cell.2009.06.015

C. Combredet, V. Labrousse, L. Mollet, C. Lorin, F. Delebecque et al., A Molecularly Cloned Schwarz Strain of Measles Virus Vaccine Induces Strong Immune Responses in Macaques and Transgenic Mice, Journal of Virology, vol.77, issue.21, pp.11546-11554, 2003.
DOI : 10.1128/JVI.77.21.11546-11554.2003

URL : https://hal.archives-ouvertes.fr/hal-02129918

M. De-veer, M. Holko, M. Frevel, E. Walker, S. Der et al., Functional classification of interferon-stimulated genes identified using microarrays, Journal of Leukocyte Biology, vol.69, pp.912-920, 2001.

S. Deddouche, D. Goubau, J. Rehwinkel, P. Chakravarty, S. Begum et al., Identification of an LGP2-associated MDA5 agonist in picornavirusinfected cells, 2014.

E. Dixit and J. Kagan, Intracellular Pathogen Detection by RIG-I-Like Receptors, Advances in Immunology, vol.117, pp.99-125, 2013.
DOI : 10.1016/B978-0-12-410524-9.00004-9

J. Errett and M. Gale, Emerging complexity and new roles for the RIG-I-like receptors in innate antiviral immunity, Virologica Sinica, vol.12, issue.3, pp.163-173, 2015.
DOI : 10.1038/ni.1979

S. Es-saad, N. Tremblay, M. Baril, and D. Lamarre, Regulators of innate immunity as novel targets for panviral therapeutics, Current Opinion in Virology, vol.2, issue.5, pp.622-628, 2012.
DOI : 10.1016/j.coviro.2012.08.009

N. Garmashova, R. Gorchakov, E. Frolova, and I. Frolov, Sindbis Virus Nonstructural Protein nsP2 Is Cytotoxic and Inhibits Cellular Transcription, Journal of Virology, vol.80, issue.12, pp.5686-5696, 2006.
DOI : 10.1128/JVI.02739-05

P. Giraudon and T. Wild, Monoclonal Antibodies Against Measles Virus, Journal of General Virology, vol.54, issue.2, pp.325-332, 1981.
DOI : 10.1099/0022-1317-54-2-325

D. Goubau, S. Deddouche, R. Sousa, and C. , Cytosolic Sensing of Viruses, Immunity, vol.38, issue.5, pp.855-869, 2013.
DOI : 10.1016/j.immuni.2013.05.007

F. Guo, J. Mead, N. Aliya, L. Wang, A. Cuconati et al., RO 90-7501 Enhances TLR3 and RLR Agonist Induced Antiviral Response, PLoS ONE, vol.7, issue.10, 2012.
DOI : 10.1371/journal.pone.0042583.g009

H. Gad, H. Paulous, S. Belarbi, E. Diancourt, L. Drosten et al., Desprè s P. 2012. The E2-E166K substitution restores Chikungunya virus growth in OAS3 expressing cells by acting on viral entry, Virology, vol.434, pp.27-37

S. Ikegame, M. Takeda, S. Ohno, Y. Nakatsu, Y. Nakanishi et al., Both RIG-I and MDA5 RNA Helicases Contribute to the Induction of Alpha/Beta Interferon in Measles Virus-Infected Human Cells, Journal of Virology, vol.84, issue.1, pp.372-37901690, 2010.
DOI : 10.1128/JVI.01690-09

B. Jagla, B. Wiswedel, and J. Coppé-e, Extending KNIME for next-generation sequencing data analysis, Bioinformatics, vol.29, issue.Suppl. 2, 2011.
DOI : 10.1038/nbt.1754

H. Kato, O. Takeuchi, E. Mikamo-satoh, R. Hirai, T. Kawai et al., Length-dependent recognition of double-stranded ribonucleic acids by retinoic acid???inducible gene-I and melanoma differentiation???associated gene 5, The Journal of Experimental Medicine, vol.89, issue.7, pp.1601-1610, 2008.
DOI : 10.1016/j.jsb.2005.05.002

A. Kell, M. Stoddard, H. Li, J. Marcotrigiano, G. Shaw et al., ABSTRACT, Journal of Virology, vol.89, issue.21, pp.11056-1106801964, 2015.
DOI : 10.1128/JVI.01964-15

A. Komarova, C. Combredet, L. Meyniel-schicklin, M. Chapelle, G. Caignard et al., Proteomic Analysis of Virus-Host Interactions in an Infectious Context Using Recombinant Viruses, Molecular & Cellular Proteomics, vol.162, issue.12, 2011.
DOI : 10.1073/pnas.0400567101

URL : https://hal.archives-ouvertes.fr/hal-00639495

A. Komarova, C. Combredet, O. Sismeiro, M. Dillies, B. Jagla et al., Identification of RNA partners of viral proteins in infected cells, RNA Biology, vol.57, issue.6, 2013.
DOI : 10.1093/bioinformatics/btp616

B. Langmead, C. Trapnell, M. Pop, and S. Salzberg, Ultrafast and memory-efficient alignment of short DNA sequences to the human genome, Genome Biology, vol.10, issue.3, pp.10-1186, 2009.
DOI : 10.1186/gb-2009-10-3-r25

H. Li, B. Handsaker, A. Wysoker, T. Fennell, J. Ruan et al., The Sequence Alignment/Map format and SAMtools, Bioinformatics, vol.9, issue.11, pp.2078-2079, 1000.
DOI : 10.1146/annurev.genom.9.081307.164359

B. Liddicoat, R. Piskol, A. Chalk, G. Ramaswami, M. Higuchi et al., RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself, Science, vol.7, issue.7, pp.1115-1120, 2015.
DOI : 10.1371/journal.pone.0040321

P. Liehl, V. Zuzarte-luís, J. Chan, T. Zillinger, F. Baptista et al., Host-cell sensors for Plasmodium activate innate immunity against liver-stage infection, Nature Medicine, vol.12, issue.1, 2014.
DOI : 10.1007/BF00932618

R. Lin, P. Gé-nin, Y. Mamane, and J. Hiscott, Selective DNA Binding and Association with the CREB Binding Protein Coactivator Contribute to Differential Activation of Alpha/Beta Interferon Genes by Interferon Regulatory Factors 3 and 7, Molecular and Cellular Biology, vol.20, issue.17, pp.6342-6353, 2000.
DOI : 10.1128/MCB.20.17.6342-6353.2000

Y. Loo and M. Gale, Immune Signaling by RIG-I-like Receptors, Immunity, vol.34, issue.5, pp.680-692, 2011.
DOI : 10.1016/j.immuni.2011.05.003

M. Love, W. Huber, and S. Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biology, vol.14, issue.12, p.550, 2014.
DOI : 10.1186/gb-2013-14-4-r36

M. Lucas-hourani, D. Dauzonne, P. Jorda, G. Cousin, A. Lupan et al., Inhibition of Pyrimidine Biosynthesis Pathway Suppresses Viral Growth through Innate Immunity, PLoS Pathogens, vol.8, issue.10, 2013.
DOI : 10.1371/journal.ppat.1003678.s014

URL : https://hal.archives-ouvertes.fr/pasteur-01113535

K. Malathi, B. Dong, M. Gale, and R. Silverman, Small self-RNA generated by RNase L amplifies antiviral innate immunity, Nature, vol.69, issue.7155, pp.816-819, 2007.
DOI : 10.1371/journal.ppat.0020025

K. Malathi, T. Saito, N. Crochet, D. Barton, M. Gale et al., RNase L releases a small RNA from HCV RNA that refolds into a potent PAMP, RNA, vol.16, issue.11, pp.2108-2119, 2010.
DOI : 10.1261/rna.2244210

A. Marriott and N. Dimmock, Defective interfering viruses and their potential as antiviral agents, Reviews in Medical Virology, vol.87, issue.1, pp.51-62, 2010.
DOI : 10.1007/978-94-011-7930-0_22

K. Monroe, S. Mcwhirter, and R. Vance, Identification of Host Cytosolic Sensors and Bacterial Factors Regulating the Type I Interferon Response to Legionella pneumophila, PLoS Pathogens, vol.41, issue.11, 2009.
DOI : 10.1371/journal.ppat.1000665.s003

E. Moresco and B. Beutler, LGP2: Positive about viral sensing, Proceedings of the National Academy of Sciences, vol.104, issue.2, pp.1261-1262, 2010.
DOI : 10.1073/pnas.0606699104

C. Motz, K. Schuhmann, A. Kirchhofer, M. Moldt, G. Witte et al., Paramyxovirus V Proteins Disrupt the Fold of the RNA Sensor MDA5 to Inhibit Antiviral Signaling, Science, vol.238, issue.2, pp.690-693, 2013.
DOI : 10.1006/viro.1997.8866

J. Patel and A. García-sastre, Activation and regulation of pathogen sensor RIG-I, Cytokine & Growth Factor Reviews, vol.25, issue.5, pp.513-523, 2014.
DOI : 10.1016/j.cytogfr.2014.08.005

A. Peisley, C. Lin, B. Wu, M. Orme-johnson, M. Liu et al., Cooperative assembly and dynamic disassembly of MDA5 filaments for viral dsRNA recognition, Proceedings of the National Academy of Sciences, vol.260, issue.28, pp.21010-21015, 2011.
DOI : 10.1251/bpo70

A. Pichlmair, O. Schulz, C. Tan, J. Rehwinkel, H. Kato et al., Activation of MDA5 Requires Higher-Order RNA Structures Generated during Virus Infection, Journal of Virology, vol.83, issue.20, pp.10761-1076900770, 2009.
DOI : 10.1128/JVI.00770-09

S. Plumet, W. Duprex, and D. Gerlier, Dynamics of Viral RNA Synthesis during Measles Virus Infection, Journal of Virology, vol.79, issue.11, pp.6900-6908, 2005.
DOI : 10.1128/JVI.79.11.6900-6908.2005

URL : https://hal.archives-ouvertes.fr/hal-00123404

P. Pushko, M. Parker, G. Ludwig, N. Davis, R. Johnston et al., Replicon-Helper Systems from Attenuated Venezuelan Equine Encephalitis Virus: Expression of Heterologous Genesin Vitroand Immunization against Heterologous Pathogensin Vivo, Virology, vol.239, issue.2, pp.389-401, 1997.
DOI : 10.1006/viro.1997.8878

J. Rehwinkel, C. Tan, D. Goubau, O. Schulz, A. Pichlmair et al., RIG-I Detects Viral Genomic RNA during Negative-Strand RNA Virus Infection, Cell, vol.140, issue.3, pp.397-408, 2010.
DOI : 10.1016/j.cell.2010.01.020

S. Runge, K. Sparrer, C. Lä-ssig, K. Hembach, A. Baum et al., In Vivo Ligands of MDA5 and RIG-I in Measles Virus-Infected Cells, PLoS Pathogens, vol.86, issue.4, 2014.
DOI : 10.1371/journal.ppat.1004081.s013

T. Saito, D. Owen, F. Jiang, J. Marcotrigiano, and M. Gale, Innate immunity induced by composition-dependent RIG-I recognition of hepatitis C virus RNA, Nature, vol.448, issue.7203, pp.523-527, 2008.
DOI : 10.1038/nature07106

S. Sato, K. Li, T. Kameyama, T. Hayashi, Y. Ishida et al., The RNA Sensor RIG-I Dually Functions as an Innate Sensor and Direct Antiviral Factor for Hepatitis B Virus, Immunity, vol.42, issue.1, pp.123-132, 2015.
DOI : 10.1016/j.immuni.2014.12.016

T. Satoh, H. Kato, Y. Kumagai, M. Yoneyama, S. Sato et al., LGP2 is a positive regulator of RIG-I- and MDA5-mediated antiviral responses, Proceedings of the National Academy of Sciences, vol.194, issue.3, pp.1512-1517, 2010.
DOI : 10.1084/jem.194.3.247

C. Schilte, T. Couderc, F. Chretien, M. Sourisseau, N. Gangneux et al., Type I IFN controls chikungunya virus via its action on nonhematopoietic cells, The Journal of Experimental Medicine, vol.58, issue.2, pp.429-442, 2010.
DOI : 10.1038/77553

URL : https://hal.archives-ouvertes.fr/pasteur-00458108

G. Schnell, Y. Loo, J. Marcotrigiano, and M. Gale, Uridine Composition of the Poly-U/UC Tract of HCV RNA Defines Non-Self Recognition by RIG-I, PLoS Pathogens, vol.11, issue.8, 2012.
DOI : 10.1371/journal.ppat.1002839.s001

I. Schuffenecker, I. Iteman, A. Michault, S. Murri, L. Frangeul et al., Genome Microevolution of Chikungunya Viruses Causing the Indian Ocean Outbreak, PLoS Medicine, vol.105, issue.7, 2006.
DOI : 10.1371/journal.pmed.0030263.st005

URL : https://hal.archives-ouvertes.fr/pasteur-01659363

M. Shingai, T. Ebihara, N. Begum, A. Kato, T. Honma et al., Differential Type I IFN-Inducing Abilities of Wild-Type versus Vaccine Strains of Measles Virus, The Journal of Immunology, vol.179, issue.9, pp.6123-6133, 2007.
DOI : 10.4049/jimmunol.179.9.6123

K. Shirey, Q. Nhu, K. Yim, Z. Roberts, J. Teijaro et al., The anti-tumor agent, 5,6-dimethylxanthenone-4-acetic acid (DMXAA), induces IFN-??-mediated antiviral activity in vitro and in vivo, Journal of Leukocyte Biology, vol.50, issue.3, pp.351-357, 2011.
DOI : 10.1086/651605

L. Strahle, D. Garcin, and D. Kolakofsky, Sendai virus defective-interfering genomes and the activation of interferon-beta, Virology, vol.351, issue.1, pp.101-111, 2006.
DOI : 10.1016/j.virol.2006.03.022

J. Strauss and E. Strauss, The alphaviruses: gene expression, replication, and evolution, Microbiological Reviews, vol.58, pp.491-562, 1994.

R. Team, R Foundation for Statistical Computing, 2013.

F. Triana-alonso, M. Dabrowski, J. Wadzack, and K. Nierhaus, Transcription with T7 RNA Polymerase, Journal of Biological Chemistry, vol.7, issue.11, pp.6298-6307, 1995.
DOI : 10.1093/nar/9.1.133

E. Vasseur, E. Patin, G. Laval, S. Pajon, S. Fornarino et al., The selective footprints of viral pressures at the human RIG-I-like receptor family, Human Molecular Genetics, vol.37, issue.22, pp.4462-4474, 2011.
DOI : 10.1093/nar/gkn664

T. Venkataraman, M. Valdes, R. Elsby, S. Kakuta, G. Caceres et al., Loss of DExD/H Box RNA Helicase LGP2 Manifests Disparate Antiviral Responses, The Journal of Immunology, vol.178, issue.10, pp.6444-6455, 2007.
DOI : 10.4049/jimmunol.178.10.6444

URL : http://www.jimmunol.org/content/178/10/6444.full.pdf

L. White, T. Sali, D. Alvarado, E. Gatti, P. Pierre et al., Chikungunya Virus Induces IPS-1-Dependent Innate Immune Activation and Protein Kinase R-Independent Translational Shutoff, Journal of Virology, vol.85, issue.1, pp.606-62000767, 2011.
DOI : 10.1128/JVI.00767-10

URL : https://jvi.asm.org/content/85/1/606.full.pdf

B. Wu, A. Peisley, C. Richards, H. Yao, X. Zeng et al., Structural Basis for dsRNA Recognition, Filament Formation, and Antiviral Signal Activation by MDA5, Cell, vol.152, issue.1-2, pp.276-289, 2013.
DOI : 10.1016/j.cell.2012.11.048

URL : https://doi.org/10.1016/j.cell.2012.11.048

M. Yoneyama, K. Onomoto, M. Jogi, T. Akaboshi, and T. Fujita, Viral RNA detection by RIG-I-like receptors, Current Opinion in Immunology, vol.32, pp.48-53, 2015.
DOI : 10.1016/j.coi.2014.12.012

H. Zhang, Z. Liu, Y. Sun, J. Zhu, S. Lu et al., Rig-I regulates NF-??B activity through binding to Nf-??b1 3'-UTR mRNA, Proceedings of the National Academy of Sciences, vol.26, issue.13, pp.6459-6464, 2013.
DOI : 10.1038/sj.emboj.7601751

URL : http://www.pnas.org/content/110/16/6459.full.pdf

Y. Zhang, B. Si, X. Kang, Y. Hu, J. Li et al., The confirmation of three repeated sequence elements in the 3??? untranslated region of Chikungunya virus, Virus Genes, vol.240, issue.10, pp.165-166, 2013.
DOI : 10.1006/viro.1997.8907

R. Zufferey, D. Nagy, R. Mandel, L. Naldini, and D. Trono, Multiply attenuated lentiviral vector achieves efficient gene delivery in vivo, Nature Biotechnology, vol.88, issue.9, pp.871-875, 1997.
DOI : 10.1038/nbt0997-871