C. Theves, E. Crubezy, and P. Biagini, History of Smallpox and Its Spread in Human Populations, Microbiol Spectr, vol.4, issue.4, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01382482

R. A. Rafeek, M. V. Divarathna, and F. Noordeen, History and current trends in influenza virus infections with special reference to Sri Lanka, vol.28, pp.225-232, 2017.

I. A. Luthy, V. Ritacco, and I. N. Kantor, Medicina (B Aires), vol.78, issue.2, pp.113-118, 2018.

A. L. Banuls, Mycobacterium tuberculosis: ecology and evolution of a human bacterium, J Med Microbiol, vol.64, issue.11, pp.1261-1270, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01949403

R. Rappuoli, From Pasteur to genomics: progress and challenges in infectious diseases, Nat Med, vol.10, issue.11, pp.1177-85, 2004.

P. M. Sharp and B. H. Hahn, Origins of HIV and the AIDS pandemic. Cold Spring Harb Perspect Med, vol.1, p.6841, 2011.

J. Saluzzo, P. V. , and J. Gonzalez, LES VIRUS ÉMERGENTS2013

D. M. Morens, G. K. Folkers, and A. S. Fauci, The challenge of emerging and re-emerging infectious diseases, Nature, vol.430, issue.6996, pp.242-251, 2004.

R. L. Graham, E. F. Donaldson, and R. S. Baric, A decade after SARS: strategies for controlling emerging coronaviruses, Nat Rev Microbiol, issue.11, pp.836-884, 2013.

D. S. Hui, Middle East respiratory syndrome coronavirus: risk factors and determinants of primary, household, and nosocomial transmission, Lancet Infect Dis, 2018.

B. Kumar, The emerging influenza virus threat: status and new prospects for its therapy and control, Arch Virol, vol.163, issue.4, pp.831-844, 2018.

F. Zhang, Characterization of pseudoparticles paired with hemagglutinin and neuraminidase from highly pathogenic H5N1 influenza and avian influenza A (H7N9) viruses, Virus Res, vol.253, pp.20-27, 2018.

Y. Poovorawan, Global alert to avian influenza virus infection: from H5N1 to H7N9. Pathog Glob Health, vol.107, pp.217-240, 2013.

D. Cantoni and J. S. Rossman, Ebolaviruses: New roles for old proteins, PLoS Negl Trop Dis, vol.12, issue.5, p.6349, 2018.

H. A. Elshabrawy, T. B. Erickson, and B. S. Prabhakar, Ebola virus outbreak, updates on current therapeutic strategies, Rev Med Virol, vol.25, issue.4, pp.241-53, 2015.

A. Bernadou, Measles outbreak linked to insufficient vaccination coverage in NouvelleAquitaine Region, Euro Surveill, vol.30, issue.23, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02155494

F. Krauer, Zika Virus Infection as a Cause of Congenital Brain Abnormalities and Guillain-Barre Syndrome: Systematic Review, PLoS Med, vol.14, issue.1, 2017.

E. De-clercq and G. Li, Approved Antiviral Drugs over the Past 50 Years, Clin Microbiol Rev, vol.29, issue.3, pp.695-747, 2016.

N. Abdel-haq, New antiviral agents, Indian J Pediatr, vol.73, issue.4, pp.313-334, 2006.

J. F. Marcelletti, Synergistic inhibition of herpesvirus replication by docosanol and antiviral nucleoside analogs, Antiviral Res, vol.56, issue.2, pp.153-66, 2002.

L. Delang, R. Abdelnabi, and J. Neyts, Favipiravir as a potential countermeasure against neglected and emerging RNA viruses, Antiviral Res, vol.153, pp.85-94, 2018.

J. A. Kim, Favipiravir and Ribavirin Inhibit Replication of Asian and African Strains of Zika Virus in Different Cell Models, Viruses, vol.10, issue.2, 2018.

D. Sissoko, Experimental Treatment with Favipiravir for Ebola Virus Disease (the JIKI Trial): A Historically Controlled, Single-Arm Proof-of-Concept Trial in Guinea, PLoS Med, vol.13, issue.3, p.1001967, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01327168

S. Pol and M. Corouge, Treatment of hepatitis C: perspectives, Med Mal Infect, vol.44, issue.10, pp.449-54, 2014.

S. M. Woollard and G. D. Kanmogne, Maraviroc: a review of its use in HIV infection and beyond, Drug Des Devel Ther, vol.9, pp.5447-68, 2015.

J. M. Pawlotsky, The science of direct-acting antiviral and host-targeted agent therapy, Antivir Ther, vol.17, issue.6, pp.1109-1126, 2012.

A. Karlas, A human genome-wide loss-of-function screen identifies effective chikungunya antiviral drugs, Nat Commun, vol.7, p.11320, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01515939

C. Gilbert, Statins could be used to control replication of some viruses, including HIV-1, Viral Immunol, vol.18, issue.3, pp.474-89, 2005.

Q. Nevers, Characterization of the Anti-Hepatitis C Virus Activity of New Nonpeptidic Small-Molecule Cyclophilin Inhibitors with the Potential for Broad Anti-Flaviviridae Activity, Antimicrob Agents Chemother, issue.7, p.62, 2018.

S. E. Jones, Human Papillomavirus DNA Methylation Predicts Response to Treatment Using Cidofovir and Imiquimod in Vulval Intraepithelial Neoplasia 3. Clin Cancer Res, vol.23, pp.5460-5468, 2017.

K. Watashi, Cyclosporin A and its analogs inhibit hepatitis B virus entry into cultured hepatocytes through targeting a membrane transporter, sodium taurocholate cotransporting polypeptide (NTCP), Hepatology, vol.59, issue.5, pp.1726-1763, 2014.

Y. Wang, Calcineurin inhibitors stimulate and mycophenolic acid inhibits replication of hepatitis E virus, Gastroenterology, vol.146, issue.7, pp.1775-83, 2014.

S. Kang, Suppressing dengue-2 infection by chemical inhibition of Aedes aegypti host factors, PLoS Negl Trop Dis, vol.8, issue.8, p.3084, 2014.

M. S. Diamond, M. Zachariah, and E. Harris, Mycophenolic acid inhibits dengue virus infection by preventing replication of viral RNA, Virology, vol.304, issue.2, pp.211-232, 2002.

S. D. Henry, Mycophenolic acid inhibits hepatitis C virus replication and acts in synergy with cyclosporin A and interferon-alpha, Gastroenterology, vol.131, issue.5, pp.1452-62, 2006.

B. J. Hart, Interferon-beta and mycophenolic acid are potent inhibitors of Middle East respiratory syndrome coronavirus in cell-based assays, J Gen Virol, vol.95, pp.571-578, 2014.

K. W. Cheng, Thiopurine analogs and mycophenolic acid synergistically inhibit the papain-like protease of Middle East respiratory syndrome coronavirus, Antiviral Res, vol.115, pp.9-16, 2015.

J. P. Martinez, Antiviral drug discovery: broad-spectrum drugs from nature, Nat Prod Rep, vol.32, issue.1, pp.29-48, 2015.

J. K. Dhanoa, Long non-coding RNA: its evolutionary relics and biological implications in mammals: a review, J Anim Sci Technol, vol.60, p.25, 2018.

S. Djebali, Landscape of transcription in human cells, Nature, vol.489, issue.7414, pp.101-109, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01216755

G. St-laurent, C. Wahlestedt, and P. Kapranov, The Landscape of long noncoding RNA classification, Trends Genet, vol.31, issue.5, pp.239-51, 2015.

T. Derrien, The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression, Genome Res, vol.22, issue.9, pp.1775-89, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01205054

M. Mele, Chromatin environment, transcriptional regulation, and splicing distinguish lincRNAs and mRNAs, Genome Res, vol.27, issue.1, pp.27-37, 2017.

X. Y. Meng, Long Non-Coding RNAs: Emerging and Versatile Regulators in Host-Virus Interactions. Front Immunol, vol.8, p.1663, 2017.

G. Samudyata, A. Castelo-branco, and . Bonetti, Birth, coming of age and death: The intriguing life of long noncoding RNAs, Semin Cell Dev Biol, vol.79, pp.143-152, 2018.

M. Niemczyk, Imprinted chromatin around DIRAS3 regulates alternative splicing of GNG12-AS1, a long noncoding RNA, Am J Hum Genet, vol.93, issue.2, pp.224-259, 2013.

J. E. Wilusz, Long noncoding RNAs: Re-writing dogmas of RNA processing and stability, Biochim Biophys Acta, vol.1859, issue.1, pp.128-166, 2016.

Y. Fang and M. J. Fullwood, Roles, Functions, and Mechanisms of Long Non-coding RNAs in Cancer, Genomics Proteomics Bioinformatics, vol.14, issue.1, pp.42-54, 2016.

Y. B. Beaulieu, Polyadenylation-dependent control of long noncoding RNA expression by the poly(A)-binding protein nuclear 1, PLoS Genet, vol.8, issue.11, p.1003078, 2012.

J. J. Quinn and H. Y. Chang, Unique features of long non-coding RNA biogenesis and function, Nat Rev Genet, vol.17, issue.1, pp.47-62, 2016.

K. W. Vance and C. P. Ponting, Transcriptional regulatory functions of nuclear long noncoding RNAs, Trends Genet, vol.30, issue.8, pp.348-55, 2014.

M. Beltran, A natural antisense transcript regulates Zeb2/Sip1 gene expression during Snail1-induced epithelial-mesenchymal transition, Genes Dev, vol.22, issue.6, pp.756-69, 2008.

C. M. Clemson, An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles, Mol Cell, vol.33, issue.6, pp.717-743, 2009.

J. H. Noh, Cytoplasmic functions of long noncoding RNAs, Wiley Interdiscip Rev RNA, vol.9, issue.3, p.1471, 2018.

D. E. Golden, V. R. Gerbasi, and E. J. Sontheimer, An inside job for siRNAs, Mol Cell, vol.31, issue.3, pp.309-321, 2008.

B. Czech, An endogenous small interfering RNA pathway in Drosophila, Nature, vol.453, issue.7196, pp.798-802, 2008.

K. C. Pang, Genome-wide identification of long noncoding RNAs in CD8+ T cells, J Immunol, vol.182, issue.12, pp.7738-7786, 2009.

D. M. Anderson, A micropeptide encoded by a putative long noncoding RNA regulates muscle performance, Cell, vol.160, issue.4, pp.595-606, 2015.

J. Z. Huang, A Peptide Encoded by a Putative lncRNA HOXB-AS3 Suppresses Colon Cancer Growth, Mol Cell, vol.68, issue.1, p.6, 2017.

O. Karlsson and A. A. Baccarelli, Environmental Health and Long Non-coding RNAs, Curr Environ Health Rep, vol.3, issue.3, pp.178-87, 2016.

M. B. Clark, Genome-wide analysis of long noncoding RNA stability, Genome Res, vol.22, issue.5, pp.885-98, 2012.

J. H. Yoon, LincRNA-p21 suppresses target mRNA translation, Mol Cell, vol.47, issue.4, pp.648-55, 2012.

J. H. Yoon, Scaffold function of long non-coding RNA HOTAIR in protein ubiquitination, Nat Commun, vol.4, p.2939, 2013.

J. H. Yoon, PAR-CLIP analysis uncovers AUF1 impact on target RNA fate and genome integrity, Nat Commun, vol.5, p.5248, 2014.

J. H. Yoon, J. Kim, and M. Gorospe, Long noncoding RNA turnover, Biochimie, vol.117, pp.15-21, 2015.

J. Ouyang, J. Hu, and J. L. Chen, lncRNAs regulate the innate immune response to viral infection, Wiley Interdiscip Rev RNA, vol.7, issue.1, pp.129-172, 2016.

H. Ma, The Long Noncoding RNA NEAT1 Exerts Antihantaviral Effects by Acting as Positive Feedback for RIG-I Signaling, J Virol, issue.9, p.91, 2017.

H. Kambara, Negative regulation of the interferon response by an interferon-induced long non-coding RNA, Nucleic Acids Res, vol.42, issue.16, pp.10668-80, 2014.

Y. Xiong, The STAT3-regulated long non-coding RNA Lethe promote the HCV replication, Biomed Pharmacother, vol.72, pp.165-71, 2015.

J. Wang, Host Long Noncoding RNA lncRNA-PAAN Regulates the Replication of Influenza A Virus, Viruses, issue.6, p.10, 2018.

H. Imam, The lncRNA NRON modulates HIV-1 replication in a NFAT-dependent manner and is differentially regulated by early and late viral proteins, vol.5, p.8639, 2015.

B. D. Clarke, Functional non-coding RNAs derived from the flavivirus 3' untranslated region, Virus Res, vol.206, pp.53-61, 2015.

W. C. Ng, The 5' and 3' Untranslated Regions of the Flaviviral Genome, 2017.

P. Fortes and K. V. Morris, Long noncoding RNAs in viral infections, Virus Res, vol.212, pp.1-11, 2016.

R. C. Lee, R. L. Feinbaum, and V. Ambros, The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14, Cell, vol.75, issue.5, pp.843-54, 1993.
URL : https://hal.archives-ouvertes.fr/in2p3-00597159

Y. Lee, MicroRNA maturation: stepwise processing and subcellular localization, EMBO J, vol.21, issue.17, pp.4663-70, 2002.

Y. Lee, MicroRNA genes are transcribed by RNA polymerase II, EMBO J, vol.23, pp.4051-60, 1920.

R. P. Kincaid, J. M. Burke, and C. S. Sullivan, RNA virus microRNA that mimics a B-cell oncomiR, Proc Natl Acad Sci, vol.109, issue.8, pp.3077-82, 2012.

K. Wu, The Role of Exportin-5 in MicroRNA Biogenesis and Cancer, Genomics Proteomics Bioinformatics, vol.16, issue.2, pp.120-126, 2018.

X. Cai, C. H. Hagedorn, and B. R. Cullen, Human microRNAs are processed from capped, polyadenylated transcripts that can also function as mRNAs, RNA, vol.10, issue.12, pp.1957-66, 2004.

V. C. Auyeung, Beyond secondary structure: primary-sequence determinants license primiRNA hairpins for processing, Cell, vol.152, issue.4, pp.844-58, 2013.

J. Han, Molecular basis for the recognition of primary microRNAs by the Drosha-DGCR8 complex, Cell, vol.125, issue.5, pp.887-901, 2006.

Y. Zeng and B. R. Cullen, Efficient processing of primary microRNA hairpins by Drosha requires flanking nonstructured RNA sequences, J Biol Chem, vol.280, issue.30, pp.27595-603, 2005.

A. Creugny, A. Fender, and S. Pfeffer, Regulation of primary microRNA processing, FEBS Lett, vol.592, issue.12, pp.1980-1996, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02294841

S. Link, S. E. Grund, and S. Diederichs, Alternative splicing affects the subcellular localization of Drosha, Nucleic Acids Res, vol.44, issue.11, pp.5330-5373, 2016.

S. C. Kwon, Structure of Human DROSHA. Cell, vol.164, issue.1-2, pp.81-90, 2016.

J. Han, The Drosha-DGCR8 complex in primary microRNA processing, Genes Dev, vol.18, issue.24, pp.3016-3043, 2004.

R. Senturia, Structure of the dimerization domain of DiGeorge critical region 8, Protein Sci, vol.19, issue.7, pp.1354-65, 2010.

Y. Zeng, R. Yi, and B. R. Cullen, Recognition and cleavage of primary microRNA precursors by the nuclear processing enzyme Drosha, EMBO J, vol.24, issue.1, pp.138-186, 2005.

M. Trabucchi, KSRP Promotes the Maturation of a Group of miRNA Precuresors, Adv Exp Med Biol, vol.700, pp.36-42, 2011.

M. T. Bohnsack, K. Czaplinski, and D. Gorlich, Exportin 5 is a RanGTP-dependent dsRNAbinding protein that mediates nuclear export of pre-miRNAs, RNA, vol.10, issue.2, pp.185-91, 2004.

E. Lund, Nuclear export of microRNA precursors, Science, vol.303, issue.5654, pp.95-103, 2004.

H. Zhang, Single processing center models for human Dicer and bacterial RNase III, Cell, vol.118, issue.1, pp.57-68, 2004.

S. Gu, The loop position of shRNAs and pre-miRNAs is critical for the accuracy of dicer processing in vivo, Cell, vol.151, issue.4, pp.900-911, 2012.

I. J. Macrae, Structural basis for double-stranded RNA processing by Dicer, Science, vol.311, issue.5758, pp.195-203, 2006.

M. Ha and V. N. Kim, Regulation of microRNA biogenesis, Nat Rev Mol Cell Biol, vol.15, issue.8, pp.509-533, 2014.

I. J. Macrae, K. Zhou, and J. A. Doudna, Structural determinants of RNA recognition and cleavage by Dicer, Nat Struct Mol Biol, vol.14, issue.10, pp.934-974, 2007.

J. E. Park, Dicer recognizes the 5' end of RNA for efficient and accurate processing, Nature, vol.475, issue.7355, pp.201-206, 2011.

T. P. Chendrimada, TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing, Nature, vol.436, issue.7051, pp.740-744, 2005.

M. Trabucchi, The RNA-binding protein KSRP promotes the biogenesis of a subset of microRNAs, Nature, vol.459, issue.7249, pp.1010-1014, 2009.

B. Xhemalce, S. C. Robson, and T. Kouzarides, Human RNA methyltransferase BCDIN3D regulates microRNA processing, Cell, vol.151, issue.2, pp.278-88, 2012.

T. Kawamata, Y. Tomari, and R. Making, Trends Biochem Sci, vol.35, issue.7, pp.368-76, 2010.

A. V. Olina, Argonaute Proteins and Mechanisms of RNA Interference in Eukaryotes and Prokaryotes, Biochemistry (Mosc), vol.83, issue.5, pp.483-497, 2018.

H. Su, Essential and overlapping functions for mammalian Argonautes in microRNA silencing, Genes Dev, vol.23, issue.3, pp.304-321, 2009.

J. J. Song, Crystal structure of Argonaute and its implications for RISC slicer activity, Science, vol.305, issue.5689, pp.1434-1441, 2004.

F. Frank, N. Sonenberg, and B. Nagar, Structural basis for 5'-nucleotide base-specific recognition of guide RNA by human AGO2, Nature, vol.465, issue.7299, pp.818-840, 2010.

A. Khvorova, A. Reynolds, and S. D. Jayasena, Functional siRNAs and miRNAs exhibit strand bias, Cell, vol.115, issue.2, pp.209-225, 2003.

D. S. Schwarz, Asymmetry in the assembly of the RNAi enzyme complex, Cell, vol.115, issue.2, pp.199-208, 2003.

H. A. Meijer, E. M. Smith, and M. Bushell, Regulation of miRNA strand selection: follow the leader?, Biochem Soc Trans, vol.42, issue.4, pp.1135-1175, 2014.

J. A. Law and S. E. Jacobsen, Establishing, maintaining and modifying DNA methylation patterns in plants and animals, Nat Rev Genet, vol.11, issue.3, pp.204-224, 2010.

A. J. Pratt and I. J. Macrae, The RNA-induced silencing complex: a versatile gene-silencing machine, J Biol Chem, vol.284, issue.27, pp.17897-901, 2009.

G. Hutvagner and P. D. Zamore, A microRNA in a multiple-turnover RNAi enzyme complex, Science, vol.297, issue.5589, pp.2056-60, 2002.

E. Berezikov, Mammalian mirtron genes, Mol Cell, vol.28, issue.2, pp.328-364, 2007.

M. Xie, Mammalian 5'-capped microRNA precursors that generate a single microRNA, Cell, vol.155, issue.7, pp.1568-80, 2013.

D. Cifuentes, A novel miRNA processing pathway independent of Dicer requires Argonaute2 catalytic activity, Science, vol.328, issue.5986, pp.1694-1702, 2010.

B. P. Towler, C. I. Jones, and S. F. Newbury, Mechanisms of regulation of mature miRNAs, Biochem Soc Trans, vol.43, issue.6, pp.1208-1222, 2015.

M. P. Gantier, Analysis of microRNA turnover in mammalian cells following Dicer1 ablation, Nucleic Acids Res, vol.39, issue.13, pp.5692-703, 2011.

O. S. Rissland, S. J. Hong, and D. P. Bartel, MicroRNA destabilization enables dynamic regulation of the miR-16 family in response to cell-cycle changes, Mol Cell, vol.43, issue.6, pp.993-1004, 2011.

T. Katoh, Selective stabilization of mammalian microRNAs by 3' adenylation mediated by the cytoplasmic poly(A) polymerase GLD-2, Genes Dev, vol.23, issue.4, pp.433-441, 2009.

M. R. Jones, Zcchc11-dependent uridylation of microRNA directs cytokine expression, Nat Cell Biol, vol.11, issue.9, pp.1157-63, 2009.

B. Yao, Defining a new role of GW182 in maintaining miRNA stability, EMBO Rep, vol.13, issue.12, pp.1102-1110, 2012.

P. Sethi and W. J. Lukiw, Micro-RNA abundance and stability in human brain: specific alterations in Alzheimer's disease temporal lobe neocortex, Neurosci Lett, vol.459, issue.2, pp.100-104, 2009.

S. Bail, Differential regulation of microRNA stability, RNA, vol.16, issue.5, pp.1032-1041, 2010.

B. P. Towler, The 3'-5' exoribonuclease Dis3 regulates the expression of specific microRNAs in Drosophila wing imaginal discs, RNA Biol, vol.12, issue.7, pp.728-769, 2015.

K. T. Kuppusamy, Let-7 family of microRNA is required for maturation and adult-like metabolism in stem cell-derived cardiomyocytes, Proc Natl Acad Sci U S A, vol.112, issue.21, pp.2785-94, 2015.

L. F. Sempere, Expression profiling of mammalian microRNAs uncovers a subset of brain-expressed microRNAs with possible roles in murine and human neuronal differentiation

, Genome Biol, vol.5, issue.3, p.13, 2004.

Y. Huang, Biological functions of microRNAs: a review, J Physiol Biochem, vol.67, issue.1, pp.129-168, 2011.

S. Sassen, E. A. Miska, and C. Caldas, MicroRNA: implications for cancer, Virchows Arch, vol.452, issue.1, pp.1-10, 2008.

L. Ma, miR-9, a MYC/MYCN-activated microRNA, regulates E-cadherin and cancer metastasis, Nat Cell Biol, vol.12, issue.3, pp.247-56, 2010.

T. Thum, MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts, Nature, vol.456, issue.7224, pp.980-984, 2008.

A. El-ouaamari, miR-375 targets 3'-phosphoinositide-dependent protein kinase-1 and regulates glucose-induced biological responses in pancreatic beta-cells, Diabetes, vol.57, issue.10, pp.2708-2725, 2008.

W. Wen, Cellular microRNA-miR-548g-3p modulates the replication of dengue virus, J Infect, vol.70, issue.6, pp.631-671, 2015.

L. Wang, MiR-342-5p suppresses coxsackievirus B3 biosynthesis by targeting the 2C-coding region, Antiviral Res, vol.93, issue.2, pp.270-279, 2012.

A. Slonchak, Human MicroRNA miR-532-5p Exhibits Antiviral Activity against West Nile Virus via Suppression of Host Genes SESTD1 and TAB3 Required for Virus Replication, J Virol, vol.90, issue.5, pp.2388-402, 2015.

A. M. Dickson and J. Wilusz, Strategies for viral RNA stability: live long and prosper, Trends Genet, vol.27, issue.7, pp.286-93, 2011.

J. M. Luna, Hepatitis C virus RNA functionally sequesters miR-122, Cell, vol.160, issue.6, pp.1099-110, 2015.

J. I. Henke, microRNA-122 stimulates translation of hepatitis C virus RNA, EMBO J, vol.27, issue.24, pp.3300-3310, 2008.

T. Masaki, miR-122 stimulates hepatitis C virus RNA synthesis by altering the balance of viral RNAs engaged in replication versus translation, Cell Host Microbe, vol.17, issue.2, pp.217-245, 2015.

T. K. Scheel, A Broad RNA Virus Survey Reveals Both miRNA Dependence and Functional Sequestration, Cell Host Microbe, vol.19, issue.3, pp.409-432, 2016.

L. Feng, C. G. Duan, and H. S. Guo, Inhibition of in vivo Slicer activity of Argonaute protein 1 by the viral 2b protein independent of its dsRNA-binding function, Mol Plant Pathol, vol.14, issue.6, pp.617-639, 2013.

S. Lee, Selective degradation of host MicroRNAs by an intergenic HCMV noncoding RNA accelerates virus production, Cell Host Microbe, vol.13, issue.6, pp.678-90, 2013.

P. Pawlica, W. N. Moss, and J. A. Steitz, Host miRNA degradation by Herpesvirus saimiri small nuclear RNA requires an unstructured interacting region, RNA, vol.22, issue.8, pp.1181-1190, 2016.

M. Deng, Hepatitis B virus mRNAs functionally sequester let-7a and enhance hepatocellular carcinoma, Cancer Lett, vol.383, issue.1, pp.62-72, 2016.

N. Sharma, miR-146a suppresses cellular immune response during Japanese encephalitis virus JaOArS982 strain infection in human microglial cells, J Neuroinflammation, vol.12, p.30, 2015.

Y. Zhang and Y. K. Li, MicroRNAs in the regulation of immune response against infections, J Zhejiang Univ Sci B, vol.14, issue.1, pp.1-7, 2013.

S. Wu, miR-146a facilitates replication of dengue virus by dampening interferon induction by targeting TRAF6, J Infect, vol.67, issue.4, pp.329-370, 2013.

J. Hou, MicroRNA-146a feedback inhibits RIG-I-dependent Type I IFN production in macrophages by targeting TRAF6, IRAK1, and IRAK2, J Immunol, vol.183, issue.3, pp.2150-2158, 2009.

B. C. Ho, Inhibition of miR-146a prevents enterovirus-induced death by restoring the production of type I interferon, Nat Commun, vol.5, p.3344, 2014.

M. Hussain, West Nile virus encodes a microRNA-like small RNA in the 3' untranslated region which up-regulates GATA4 mRNA and facilitates virus replication in mosquito cells, Nucleic Acids Res, vol.40, issue.5, pp.2210-2233, 2012.

P. Bruscella, Viruses and miRNAs: More Friends than Foes. Front Microbiol, vol.8, p.824, 2017.

X. Lei, Regulation of NF-kappaB inhibitor IkappaBalpha and viral replication by a KSHV microRNA, Nat Cell Biol, vol.12, issue.2, pp.193-202, 2010.

J. Qiu and D. A. Thorley-lawson, EBV microRNA BART 18-5p targets MAP3K2 to facilitate persistence in vivo by inhibiting viral replication in B cells, Proc Natl Acad Sci, vol.111, issue.30, pp.11157-62, 2014.

Y. J. Jung, MicroRNA miR-BART20-5p stabilizes Epstein-Barr virus latency by directly targeting BZLF1 and BRLF1, J Virol, vol.88, issue.16, pp.9027-9064, 2014.

L. M. Hook, Cytomegalovirus miRNAs target secretory pathway genes to facilitate formation of the virion assembly compartment and reduce cytokine secretion, Cell Host Microbe, vol.15, issue.3, pp.363-73, 2014.

D. Kaul, A. Ahlawat, and S. D. Gupta, HIV-1 genome-encoded hiv1-mir-H1 impairs cellular responses to infection, Mol Cell Biochem, vol.323, issue.1-2, pp.143-151, 2009.

C. M. O'connor, J. Vanicek, and E. A. Murphy, Host microRNA regulation of human cytomegalovirus immediate early protein translation promotes viral latency, J Virol, vol.88, issue.10, pp.5524-5556, 2014.

H. Liang, The origin, function, and diagnostic potential of extracellular microRNAs in human body fluids, Wiley Interdiscip Rev RNA, vol.5, issue.2, pp.285-300, 2014.

C. H. Lee, J. H. Kim, and S. W. Lee, The Role of MicroRNA in Pathogenesis and as Markers of HCV Chronic Infection, Curr Drug Targets, vol.18, issue.7, pp.756-765, 2017.

D. M. Pegtel, Functional delivery of viral miRNAs via exosomes, Proc Natl Acad Sci, vol.107, issue.14, pp.6328-6361, 2010.

J. Louten, MicroRNAs Expressed during Viral Infection: Biomarker Potential and Therapeutic Considerations, Biomark Insights, issue.10, pp.25-52, 2015.

T. W. Hoffmann, G. Duverlie, and A. Bengrine, MicroRNAs and hepatitis C virus: toward the end of miR-122 supremacy, Virol J, vol.9, p.109, 2012.

L. F. Gebert, Miravirsen (SPC3649) can inhibit the biogenesis of miR-122, Nucleic Acids Res, vol.42, issue.1, pp.609-630, 2014.

M. B. Maepa, Sustained Inhibition of HBV Replication In Vivo after Systemic Injection of AAVs Encoding Artificial Antiviral Primary MicroRNAs, Mol Ther Nucleic Acids, vol.7, pp.190-199, 2017.

M. Tan, The microRNA-let-7b-mediated attenuated strain of influenza A (H1N1) virus in a mouse model, J Infect Dev Ctries, vol.10, issue.9, pp.973-981, 2016.

R. E. Drury, D. O'connor, and A. J. Pollard, The Clinical Application of MicroRNAs in Infectious Disease. Front Immunol, vol.8, p.1182, 2017.

P. Briata, KSRP, many functions for a single protein, Front Biosci, vol.16, pp.1787-96, 2011.

D. Hollingworth, KH domains with impaired nucleic acid binding as a tool for functional analysis, Nucleic Acids Res, vol.40, issue.14, pp.6873-86, 2012.

R. Gherzi, KSRP controls pleiotropic cellular functions. Semin Cell Dev Biol, vol.34, pp.2-8, 2014.

M. F. Garcia-mayoral, The structure of the C-terminal KH domains of KSRP reveals a noncanonical motif important for mRNA degradation, Structure, vol.15, issue.4, pp.485-98, 2007.

P. Briata, Functional and molecular insights into KSRP function in mRNA decay, Biochim Biophys Acta, pp.689-94, 2013.

R. Gherzi, The RNA-binding protein KSRP promotes decay of beta-catenin mRNA and is inactivated by PI3K-AKT signaling, PLoS Biol, vol.5, issue.1, p.5, 2006.

X. Zhang, The ATM kinase induces microRNA biogenesis in the DNA damage response, Mol Cell, vol.41, issue.4, pp.371-83, 2011.

P. Briata, p38-dependent phosphorylation of the mRNA decay-promoting factor KSRP controls the stability of select myogenic transcripts, Mol Cell, vol.20, issue.6, pp.891-903, 2005.

H. Yuan, SUMO1 modification of KHSRP regulates tumorigenesis by preventing the TL-G-Rich miRNA biogenesis. Mol Cancer, vol.16, p.157, 2017.

H. Min, A new regulatory protein, KSRP, mediates exon inclusion through an intronic splicing enhancer, Genes Dev, vol.11, issue.8, pp.1023-1059, 1997.

A. Russo, Autoregulatory circuit of human rpL3 expression requires hnRNP H1, NPM and KHSRP, Nucleic Acids Res, vol.39, issue.17, pp.7576-85, 2011.

A. Douablin, HnRNP A1 tethers KSRP to an exon splicing silencer that inhibits an erythroid-specific splicing event in PU.1-induced erythroleukemia, Am J Cancer Res, vol.5, issue.4, pp.1410-1432, 2015.

R. Gherzi, A KH domain RNA binding protein, KSRP, promotes ARE-directed mRNA turnover by recruiting the degradation machinery, Mol Cell, vol.14, issue.5, pp.571-83, 2004.

T. Bakheet, E. Hitti, and K. S. Khabar, ARED-Plus: an updated and expanded database of AUrich element-containing mRNAs and pre-mRNAs, Nucleic Acids Res, vol.46, issue.D1, pp.218-220, 2018.

X. Li, KSRP: a checkpoint for inflammatory cytokine production in astrocytes, Glia, vol.60, issue.11, pp.1773-84, 2012.

C. W. Bird, KSRP modulation of GAP-43 mRNA stability restricts axonal outgrowth in embryonic hippocampal neurons, PLoS One, vol.8, issue.11, p.79255, 2013.

G. Nicastro, Noncanonical G recognition mediates KSRP regulation of let-7 biogenesis, Nat Struct Mol Biol, vol.19, issue.12, pp.1282-1288, 2012.

E. Repetto, Let-7b/c enhance the stability of a tissue-specific mRNA during mammalian organogenesis as part of a feedback loop involving KSRP, PLoS Genet, vol.8, issue.7, p.1002823, 2012.

A. Gupta, MicroRNAs, hepatitis C virus, and HCV/HIV-1 co-infection: new insights in pathogenesis and therapy, Viruses, vol.4, issue.11, pp.2485-513, 2012.

Y. Y. Lin, KSRP and MicroRNA 145 are negative regulators of lipolysis in white adipose tissue, Mol Cell Biol, vol.34, issue.12, pp.2339-2388, 2014.

L. Tong, KH-type splicing regulatory protein (KHSRP) contributes to tumorigenesis by promoting miR-26a maturation in small cell lung cancer, Mol Cell Biochem, vol.422, issue.1-2, pp.61-74, 2016.

D. Pruksakorn, Overexpression of KH-type splicing regulatory protein regulates proliferation, migration, and implantation ability of osteosarcoma, Int J Oncol, vol.49, issue.3, pp.903-915, 2016.

S. Bhattacharyya, Regulation of miR-155 biogenesis in cystic fibrosis lung epithelial cells: antagonistic role of two mRNA-destabilizing proteins, KSRP and TTP, Biochem Biophys Res Commun, vol.433, issue.4, pp.484-492, 2013.

P. Briata, PI3K/AKT signaling determines a dynamic switch between distinct KSRP functions favoring skeletal myogenesis, Cell Death Differ, vol.19, issue.3, pp.478-87, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00675790

M. Giovarelli, H19 long noncoding RNA controls the mRNA decay promoting function of KSRP, Proc Natl Acad Sci, vol.111, issue.47, pp.5023-5031, 2014.

Y. Gao, The H19/let-7 double-negative feedback loop contributes to glucose metabolism in muscle cells, Nucleic Acids Res, vol.42, issue.22, pp.13799-811, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-01083965

S. Soonthornvacharin, Systems-based analysis of RIG-I-dependent signalling identifies KHSRP as an inhibitor of RIG-I receptor activation, Nat Microbiol, vol.2, p.17022, 2017.

L. L. Chen, Enterovirus 71 infection cleaves a negative regulator for viral internal ribosomal entry site-driven translation, J Virol, vol.87, issue.7, pp.3828-3866, 2013.

A. L. Liu, Comparative analysis of selected innate immune-related genes following infection of immortal DF-1 cells with highly pathogenic (H5N1) and low pathogenic (H9N2) avian influenza viruses, Virus Genes, vol.50, issue.2, pp.189-99, 2015.

W. J. Lin, Posttranscriptional control of type I interferon genes by KSRP in the innate immune response against viral infection, Mol Cell Biol, vol.31, issue.16, pp.3196-207, 2011.

F. Chai, Subcellular quantitative proteomic analysis reveals host proteins involved in human cytomegalovirus infection, Biochim Biophys Acta, vol.1854, issue.8, pp.967-78, 2015.

Q. L. Choo, Isolation of a cDNA clone derived from a blood-borne non-A, non-B viral hepatitis genome, Science, vol.244, issue.4902, pp.359-62, 1989.

J. M. Llovet, Hepatocellular carcinoma. Nat Rev Dis Primers, vol.2, p.16018, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01910322

J. C. Nault, Telomerase reverse transcriptase promoter mutation is an early somatic genetic alteration in the transformation of premalignant nodules in hepatocellular carcinoma on cirrhosis, Hepatology, vol.60, issue.6, pp.1983-92, 2014.

E. Piver, Ultrastructural organisation of HCV from the bloodstream of infected patients revealed by electron microscopy after specific immunocapture, Gut, vol.66, issue.8, pp.1487-1495, 2017.

V. Pene, Role of cleavage at the core-E1 junction of hepatitis C virus polyprotein in viral morphogenesis, PLoS One, vol.12, issue.4, p.175810, 2017.

D. Paul, V. Madan, and R. Bartenschlager, Hepatitis C virus RNA replication and assembly: living on the fat of the land, Cell Host Microbe, vol.16, issue.5, pp.569-79, 2014.

C. L. Chen, Hepatitis C virus has a genetically determined lymphotropism through coreceptor B7.2, Nat Commun, vol.8, p.13882, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01911337

J. T. Blackard, N. Kemmer, and K. E. Sherman, Extrahepatic replication of HCV: insights into clinical manifestations and biological consequences, Hepatology, vol.44, issue.1, pp.15-22, 2006.

E. Blanchard, Hepatitis C virus entry depends on clathrin-mediated endocytosis, J Virol, vol.80, issue.14, pp.6964-72, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00105504

G. Sharma, H. Raheja, and S. Das, Hepatitis C virus: Enslavement of host factors, IUBMB Life, vol.70, issue.1, pp.41-49, 2018.

K. Gawlik and P. A. Gallay, HCV core protein and virus assembly: what we know without structures, Immunol Res, vol.60, issue.1, pp.1-10, 2014.

P. Andre, Characterization of low-and very-low-density hepatitis C virus RNAcontaining particles, J Virol, vol.76, issue.14, pp.6919-6947, 2002.

M. P. Manns, Hepatitis C virus infection. Nat Rev Dis Primers, vol.3, p.17006, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01669250

J. J. Feld, Ribavirin revisited in the era of direct-acting antiviral therapy for hepatitis C virus infection, Liver Int, vol.37, issue.1, pp.5-18, 2017.

J. M. Pawlotsky, Hepatitis C Drugs: Is Next Generation the Last Generation? Gastroenterology, vol.151, pp.587-90, 2016.

L. Puchades-renau and M. Berenguer, Introduction to hepatitis C virus infection: Overview and history of hepatitis C virus therapies, Hemodial Int, vol.22, issue.1, pp.8-21, 2018.

T. Fukuhara and Y. Matsuura, Role of miR-122 and lipid metabolism in HCV infection, J Gastroenterol, vol.48, issue.2, pp.169-76, 2013.

C. L. Jopling, Modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA, Science, vol.309, issue.5740, pp.1577-81, 2005.

T. Shimakami, Base pairing between hepatitis C virus RNA and microRNA 122 3' of its seed sequence is essential for genome stabilization and production of infectious virus, J Virol, vol.86, issue.13, pp.7372-83, 2012.

G. Borgia, The therapeutic potential of new investigational hepatitis C virus translation inhibitors, Expert Opin Investig Drugs, vol.25, issue.10, pp.1209-1223, 2016.

Y. Murakami, Regulation of the hepatitis C virus genome replication by miR-199a, J Hepatol, vol.50, issue.3, pp.453-60, 2009.

S. Bandyopadhyay, Hepatitis C virus infection and hepatic stellate cell activation downregulate miR-29: miR-29 overexpression reduces hepatitis C viral abundance in culture, J Infect Dis, vol.203, issue.12, pp.1753-62, 2011.

A. C. Panda, Identification of senescence-associated circular RNAs (SAC-RNAs) reveals senescence suppressor CircPVT1, Nucleic Acids Res, vol.45, issue.7, pp.4021-4035, 2017.

M. R. Higgs, H. Lerat, and J. M. Pawlotsky, Hepatitis C virus-induced activation of beta-catenin promotes c-Myc expression and a cascade of pro-carcinogenetic events, Oncogene, vol.32, issue.39, pp.4683-93, 2013.

Z. Q. Li, Hepatitis C virus core protein impairs metabolic disorder of liver cell via HOTAIR-Sirt1 signalling, Biosci Rep, vol.36, issue.3, 2016.

X. Qian, Long non-coding RNA GAS5 inhibited hepatitis C virus replication by binding viral NS3 protein, Virology, vol.492, pp.155-65, 2016.

N. Kin and A. V. , Les infections à coronavirus humains. Revue Francophone des Laboratoires, pp.25-33, 2016.

A. Vabret,

, Pathol Biol (Paris), vol.57, issue.2, pp.149-60, 2009.

Y. Guan, Isolation and characterization of viruses related to the SARS coronavirus from animals in southern China, Science, vol.302, issue.5643, pp.276-284, 2003.

K. Bohmwald, Neurologic Alterations Due to Respiratory Virus Infections. Front Cell Neurosci, vol.12, p.386, 2018.

J. Zhou, Middle East respiratory syndrome coronavirus infection: virus-host cell interactions and implications on pathogenesis, Virol J, vol.12, p.218, 2015.

P. S. Masters, The molecular biology of coronaviruses, Adv Virus Res, vol.66, pp.193-292, 2006.

S. Perlman and J. Netland, Coronaviruses post-SARS: update on replication and pathogenesis, Nat Rev Microbiol, vol.7, issue.6, pp.439-50, 2009.

A. R. Fehr and S. Perlman, Coronaviruses: an overview of their replication and pathogenesis, Methods Mol Biol, vol.1282, pp.1-23, 2015.

E. De-wit, SARS and MERS: recent insights into emerging coronaviruses, Nat Rev Microbiol, vol.14, issue.8, pp.523-557, 2016.

A. H. De-wilde, Cyclophilins and cyclophilin inhibitors in nidovirus replication, Virology, vol.522, pp.46-55, 2018.

J. Cao, J. C. Forrest, and X. Zhang, A screen of the NIH Clinical Collection small molecule library identifies potential anti-coronavirus drugs, Antiviral Res, vol.114, pp.1-10, 2015.

M. Khalid, Ribavirin and interferon-alpha2b as primary and preventive treatment for Middle East respiratory syndrome coronavirus: a preliminary report of two cases, Antivir Ther, vol.20, issue.1, pp.87-91, 2015.

S. Lee, H. W. Kim, and K. H. Kim, Antibodies against Hepatitis A and Hepatitis B Virus in Intravenous Immunoglobulin Products, J Korean Med Sci, vol.31, issue.12, pp.1937-1942, 2016.

S. Xia, Peptide-Based Membrane Fusion Inhibitors Targeting HCoV-229E Spike Protein HR1 and HR2 Domains, Int J Mol Sci, vol.19, issue.2, 2018.

L. Cui, The Nucleocapsid Protein of Coronaviruses Acts as a Viral Suppressor of RNA Silencing in Mammalian Cells, J Virol, vol.89, issue.17, pp.9029-9072, 2015.

F. W. Lai, Human coronavirus OC43 nucleocapsid protein binds microRNA 9 and potentiates NF-kappaB activation, J Virol, vol.88, issue.1, pp.54-65, 2014.

X. Zhao, miR-27b attenuates apoptosis induced by transmissible gastroenteritis virus (TGEV) infection via targeting runt-related transcription factor 1 (RUNX1), PeerJ, vol.4, p.1635, 2016.

Y. Ma, The Coronavirus Transmissible Gastroenteritis Virus Evades the Type I Interferon Response through IRE1alpha-Mediated Manipulation of the MicroRNA miR

, Axis. J Virol, issue.22, p.92, 2018.

X. Ma, Differentially expressed non-coding RNAs induced by transmissible gastroenteritis virus potentially regulate inflammation and NF-kappaB pathway in porcine intestinal epithelial cell line, BMC Genomics, vol.19, issue.1, p.747, 2018.

L. Morales, SARS-CoV-Encoded Small RNAs Contribute to Infection-Associated Lung Pathology, Cell Host Microbe, vol.21, issue.3, pp.344-355, 2017.

A. Kaul, Cell culture adaptation of hepatitis C virus and in vivo viability of an adapted variant, J Virol, vol.81, issue.23, pp.13168-79, 2007.

C. Boukadida, NS2 proteins of GB virus B and hepatitis C virus share common protease activities and membrane topologies, J Virol, vol.88, issue.13, pp.7426-7470, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01005500

F. Liu, Efficient Suppression of Hepatitis C Virus Replication by Combination Treatment with miR-122 Antagonism and Direct-acting Antivirals in Cell Culture Systems, vol.6, p.30939, 2016.

H. Colman, Genome-wide analysis of host mRNA translation during hepatitis C virus infection, J Virol, vol.87, issue.12, pp.6668-77, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01702821

I. Diaz-moreno, Phosphorylation-mediated unfolding of a KH domain regulates KSRP localization via 14-3-3 binding, Nat Struct Mol Biol, vol.16, issue.3, pp.238-284, 2009.

R. Hou, SLC14A1: a novel target for human urothelial cancer, Clin Transl Oncol, vol.19, issue.12, pp.1438-1446, 2017.

H. An, Inscuteable maintains type I neuroblast lineage identity via Numb/Notch signaling in the Drosophila larval brain, J Genet Genomics, vol.44, issue.3, pp.151-162, 2017.

M. R. Sutherland, W. Ruf, and E. L. , Tissue factor and glycoprotein C on herpes simplex virus type 1 are protease-activated receptor 2 cofactors that enhance infection, Blood, vol.119, issue.15, pp.3638-3683, 2012.

K. Khoufache, Protective role for protease-activated receptor-2 against influenza virus pathogenesis via an IFN-gamma-dependent pathway, J Immunol, vol.182, issue.12, pp.7795-802, 2009.

A. Weithauser, Protease-activated receptor-2 regulates the innate immune response to viral infection in a coxsackievirus B3-induced myocarditis, J Am Coll Cardiol, vol.62, pp.1737-1782, 2013.

C. C. Yu, Medicine (Baltimore), KCNN2 polymorphisms and cardiac tachyarrhythmias, vol.95, p.4312, 2016.

R. S. Freeman, NGF deprivation-induced gene expression: after ten years, where do we stand? Prog Brain Res, vol.146, pp.111-137, 2004.

N. Begum, W. Shen, and V. Manganiello, Role of PDE3A in regulation of cell cycle progression in mouse vascular smooth muscle cells and oocytes: implications in cardiovascular diseases and infertility, Curr Opin Pharmacol, vol.11, issue.6, pp.725-734, 2011.

X. H. Liang, Non-coding RNA LINC00473 mediates decidualization of human endometrial stromal cells in response to cAMP signaling, vol.6, p.22744, 2016.

A. Serretti, A. Drago, and D. De-ronchi, HTR2A gene variants and psychiatric disorders: a review of current literature and selection of SNPs for future studies, Curr Med Chem, vol.14, pp.2053-69, 2007.

Y. Bao, Expression and evolutionary conservation of the tescalcin gene during development, Gene Expr Patterns, vol.9, issue.5, pp.273-81, 2009.

Y. Wang, Long non-coding RNA LINC00161 sensitises osteosarcoma cells to cisplatininduced apoptosis by regulating the miR-645-IFIT2 axis, Cancer Lett, vol.382, issue.2, pp.137-146, 2016.

F. Sanchis-gomar, Mitochondrial biogenesis in health and disease. Molecular and therapeutic approaches, Curr Pharm Des, vol.20, issue.35, pp.5619-5652, 2014.

R. E. Shalaby, PGC1alpha Transcriptional Adaptor Function Governs Hepatitis B Virus Replication by Controlling HBcAg/p21 Protein-Mediated Capsid Formation, J Virol, p.91, 1920.

C. Bernsmeier, Hepatitis C virus dysregulates glucose homeostasis by a dual mechanism involving induction of PGC1alpha and dephosphorylation of FoxO1, J Viral Hepat, vol.21, issue.1, pp.9-18, 2014.

J. Quantius, Influenza Virus Infects Epithelial Stem/Progenitor Cells of the Distal Lung: Impact on Fgfr2b-Driven Epithelial Repair, PLoS Pathog, vol.12, issue.6, p.1005544, 2016.

K. Mizuta, TMEM100: A Novel Intracellular Transmembrane Protein Essential for Vascular Development and Cardiac Morphogenesis, in Etiology and Morphogenesis of Congenital Heart Disease: From Gene Function and Cellular Interaction to Morphology, pp.169-170, 2016.

H. J. Weng, Tmem100 Is a Regulator of TRPA1-TRPV1 Complex and Contributes to Persistent Pain, Neuron, vol.85, issue.4, pp.833-879, 2015.

K. Saigusa, RGC32, a novel p53-inducible gene, is located on centrosomes during mitosis and results in G2/M arrest, vol.26, pp.1110-1131, 2007.

M. Pistoni, Chromatin association and regulation of rDNA transcription by the Rasfamily protein RasL11a, EMBO J, vol.29, issue.7, pp.1215-1239, 2010.

R. Sanuki, Panky, a novel photoreceptor-specific ankyrin repeat protein, is a transcriptional cofactor that suppresses CRX-regulated photoreceptor genes, FEBS Lett, vol.584, issue.4, pp.753-761, 2010.

A. Spaziani, Role of p38 MAPK and RNA-dependent protein kinase (PKR) in hepatitis C virus core-dependent nuclear delocalization of cyclin B1, J Biol Chem, vol.281, issue.16, pp.10983-10992, 2006.

C. K. Lai, Hepatitis C virus NS3/4A protein interacts with ATM, impairs DNA repair and enhances sensitivity to ionizing radiation, Virology, vol.370, issue.2, pp.295-309, 2008.

S. M. Shawki, Increased DNA damage in hepatitis C virus-related hepatocellular carcinoma, DNA Cell Biol, vol.33, issue.12, pp.884-90, 2014.

I. Diaz-moreno, Orientation of the central domains of KSRP and its implications for the interaction with the RNA targets, Nucleic Acids Res, vol.38, issue.15, pp.5193-205, 2010.

W. Liu and C. Ding, Roles of LncRNAs in Viral Infections. Front Cell Infect Microbiol, vol.7, p.205, 2017.

A. S. Bayoumi, Crosstalk between Long Noncoding RNAs and MicroRNAs in Health and Disease, Int J Mol Sci, vol.17, issue.3, p.356, 2016.

V. Madan, Inhibition of HCV replication by cyclophilin antagonists is linked to replication fitness and occurs by inhibition of membranous web formation, Gastroenterology, vol.146, issue.5, pp.1-9, 2014.