P. Abbink, R. A. Larocca, D. L. Barrera, R. A. Bricault, and C. A. , Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys, Science, vol.353, pp.1129-1132, 2016.

H. J. Abu-toamih-atamni, Y. Ziner, R. Mott, L. Wolf, and F. A. Iraqi, Glucose tolerance female-specific QTL mapped in collaborative cross mice, Mamm Genome, vol.28, pp.20-30, 2017.

A. Waldorf, K. M. Nelson, B. R. Stencel-baerenwald, J. E. Studholme, and C. , Congenital Zika virus infection as a silent pathology with loss of neurogenic output in the fetal brain, Nature medicine, vol.24, pp.368-374, 2018.

A. Waldorf, K. M. Stencel-baerenwald, J. E. Kapur, R. P. Studholme, and C. , Fetal brain lesions after subcutaneous inoculation of Zika virus in a pregnant nonhuman primate, Nature medicine, vol.22, pp.1256-1259, 2016.

J. J. Adibi, E. T. Marques, J. Cartus, A. Beigi, and R. H. , Teratogenic effects of the Zika virus and the role of the placenta, Lancet, vol.387, pp.1587-1590, 2016.

S. Aguirre, A. M. Maestre, S. Pagni, and J. R. Patel, DENV inhibits type I IFN production in infected cells by cleaving human STING, PLoS pathogens, vol.8, p.1002934, 2012.

M. Aid, P. Abbink, R. A. Larocca, and M. Boyd, Zika Virus Persistence in the Central Nervous System and Lymph Nodes of Rhesus Monkeys, Cell, vol.169, pp.610-620, 2017.

A. Akhtar, The flaws and human harms of animal experimentation, CQ : the international journal of healthcare ethics committees, vol.24, pp.407-419, 2015.

K. Akiyama, A. Alberdi, W. Alef, and K. Asada, First M87 Event Horizon Telescope Results. I. The Shadow of the Supermassive Black Hole, Astrophysical Journal Letters, vol.875, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02404868

M. T. Alera, L. Hermann, I. A. Tac-an, and C. Klungthong, Zika virus infection, vol.21, pp.722-724, 2012.

M. T. Aliota, E. A. Caine, E. C. Walker, K. E. Larkin, E. Camacho et al., Characterization of Lethal Zika Virus Infection in AG129 Mice, PLoS neglected tropical diseases, vol.10, p.4682, 2016.

M. Altfeld, M. M. Addo, E. S. Rosenberg, and F. M. Hecht, Influence of HLA-B57 on clinical presentation and viral control during acute HIV-1 infection, AIDS, vol.17, pp.2581-2591, 2003.

A. Santos, E. Fink, and K. , Animal Models for Dengue and Zika Vaccine Development, Advances in experimental medicine and biology 1062, pp.215-239, 2018.

L. V. Alves, C. E. Paredes, G. C. Silva, J. G. Mello, and J. G. Alves, Neurodevelopment of 24 children born in Brazil with congenital Zika syndrome in 2015: a case series study, BMJ open, vol.8, p.21304, 2018.

P. An, G. Bleiber, P. Duggal, and G. Nelson, APOBEC3G genetic variants and their influence on the progression to AIDS, Journal of virology, vol.78, pp.11070-11076, 2004.

F. Anfasa, J. Y. Siegers, M. Van-der-kroeg, and N. Mumtaz, Phenotypic Differences between Asian and African Lineage Zika Viruses in Human Neural Progenitor Cells, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01890219

N. Arpaia, J. A. Green, B. Moltedo, and A. Arvey, A Distinct Function of Regulatory T Cells in Tissue Protection, Cell, vol.162, pp.1078-1089, 2015.

D. Artis and H. Spits, The biology of innate lymphoid cells, Nature, vol.517, pp.293-301, 2015.

D. G. Ashbrook, D. Arends, P. Prins, and M. K. Mulligan, The expanded BXD family of mice: A cohort for experimental systems genetics and precision medicine, 2019.

S. Ashkar, G. F. Weber, V. Panoutsakopoulou, and M. E. Sanchirico, Eta-1 (osteopontin): an early component of type-1 (cell-mediated) immunity, Science, vol.287, pp.860-864, 2000.

J. Ashour, J. Morrison, M. Laurent-rolle, and A. Belicha-villanueva, Mouse STAT2 restricts early dengue virus replication, Cell host & microbe, vol.8, pp.410-421, 2010.

H. J. Atamni, M. Botzman, R. Mott, I. Gat-viks, and F. A. Iraqi, Mapping liver fat female-dependent quantitative trait loci in collaborative cross mice, Mamm Genome, vol.27, pp.565-573, 2016.

M. Aubry, A. Teissier, M. Huart, and S. Merceron, Emerging infectious diseases, vol.23, pp.669-672, 2014.

J. Auwerx, P. Avner, R. Baldock, and A. Ballabio, The European dimension for the mouse genome mutagenesis program, Nature genetics, vol.36, pp.925-927, 2004.

N. V. Ayala-nunez, T. E. Hoornweg, D. P. Van-de-pol, K. A. Sjollema, J. Flipse et al., How antibodies alter the cell entry pathway of dengue virus particles in macrophages, Scientific reports, vol.6, p.28768, 2016.

G. Bachelard, Le nouvel esprit scientifique, 1934.

A. Balfour, The Wild Monkey as a Reservoir for the Virus of Yellow Fever, The Lancet, vol.183, pp.1176-1178, 1914.

S. V. Bardina, P. Bunduc, S. Tripathi, and J. Duehr, Enhancement of Zika virus pathogenesis by preexisting antiflavivirus immunity, Science, vol.356, pp.175-180, 2017.

M. L. Barjas-castro, R. N. Angerami, M. S. Cunha, and A. Suzuki, Probable transfusion-transmitted Zika virus in Brazil, Transfusion, vol.56, pp.1684-1688, 2016.

K. Baruch, A. Deczkowska, E. David, and J. M. Castellano, Aging. Aging-induced type I interferon response at the choroid plexus negatively affects brain function, Science, vol.346, pp.89-93, 2014.

L. Barzon, E. Percivalle, M. Pacenti, and F. Rovida, Virus and Antibody Dynamics in Travelers With Acute Zika Virus Infection, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America, vol.66, pp.1173-1180, 2018.

D. Baud, D. J. Gubler, B. Schaub, M. C. Lanteri, and D. Musso, An update on Zika virus infection, Lancet, vol.390, pp.2099-2109, 2017.

J. T. Beaver, N. Lelutiu, R. Habib, and I. Skountzou, Evolution of Two Major Zika Virus Lineages: Implications for Pathology, Immune Response, and Vaccine Development, Front Immunol, vol.9, p.1640, 2018.

M. Besnard, S. Lastere, A. Teissier, V. Cao-lormeau, and D. Musso, Euro surveillance : bulletin Europeen sur les maladies transmissibles, European communicable disease bulletin, p.19, 2013.

B. Beutler, P. Georgel, S. Rutschmann, Z. Jiang, B. Croker et al., Genetic analysis of innate resistance to mouse cytomegalovirus (MCMV), Briefings in functional genomics & proteomics, vol.4, pp.203-213, 2005.

S. Bhatt, P. W. Gething, O. J. Brady, and J. P. Messina, The global distribution and burden of dengue, Nature, vol.496, pp.504-507, 2013.

A. M. Bingham, M. Cone, V. Mock, L. Heberlein-larson, D. Stanek et al., Comparison of Test Results for Zika Virus RNA in Urine, Serum, and Saliva Specimens from Persons with Travel-Associated Zika Virus Disease -Florida, MMWR. Morbidity and mortality weekly report, vol.65, pp.475-478, 2016.

G. M. Blohm, J. A. Lednicky, M. Marquez, and S. K. White, Evidence for Mother-to-Child Transmission of Zika Virus Through Breast Milk, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America, vol.66, pp.1120-1121, 2018.

A. Bose, F. Mouton-liger, C. Paquet, P. Mazot, M. Vigny et al., Modulation of tau phosphorylation by the kinase PKR: implications in Alzheimer's disease, Brain pathology, vol.21, pp.189-200, 2011.

D. Bottomly, M. T. Ferris, L. D. Aicher, and E. Rosenzweig, Expression quantitative trait Loci for extreme host response to influenza a in pre-collaborative cross mice, G3 (Bethesda), vol.2, pp.213-221, 2012.

J. R. Bowen, K. M. Quicke, M. S. Maddur, O. Neal, and J. T. , Zika Virus Antagonizes Type I Interferon Responses during Infection of Human Dendritic Cells, PLoS pathogens, vol.13, p.1006164, 2017.

T. Boyer-chammard, K. Schepers, S. Breurec, and T. Messiaen, Severe Thrombocytopenia after Zika Virus Infection, Emerging infectious diseases, vol.23, pp.696-698, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01520126

O. J. Brady, A. Osgood-zimmerman, N. J. Kassebaum, and S. E. Ray, The association between Zika virus infection and microcephaly in Brazil 2015-2017: An observational analysis of over 4 million births, PLoS medicine, vol.16, p.1002755, 2019.

P. Brasil, J. P. Pereira, J. Moreira, M. E. , R. Nogueira et al., Zika Virus Infection in Pregnant Women in Rio de Janeiro, N Engl J Med, vol.375, pp.2321-2334, 2016.

A. L. Brass, I. C. Huang, Y. Benita, and S. P. John, The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and dengue virus, Cell, vol.139, pp.1243-1254, 2009.

M. L. Brito-ferreira, A. De-brito, C. A. Moreira, A. De-morais-machado, and M. I. , Guillain-Barre Syndrome, Acute Disseminated Encephalomyelitis and Encephalitis Associated with Zika Virus Infection in Brazil: Detection of Viral RNA and Isolation of Virus during Late Infection, The American journal of tropical medicine and hygiene, vol.97, pp.1405-1409, 2017.

R. Buathong, L. Hermann, B. Thaisomboonsuk, and W. Rutvisuttinunt, Detection of Zika Virus Infection in Thailand, The American journal of tropical medicine and hygiene, vol.93, pp.380-383, 2015.

D. Burgner, S. E. Jamieson, and J. M. Blackwell, Genetic susceptibility to infectious diseases: big is beautiful, but will bigger be even better? The Lancet. Infectious diseases, vol.6, pp.653-663, 2006.

T. Burt, K. Yoshida, G. Lappin, and L. Vuong, Microdosing and Other Phase 0 Clinical Trials: Facilitating Translation in Drug Development, Clinical and translational science, vol.9, pp.74-88, 2016.

G. Caignard, M. M. Eva, R. Van-bruggen, and R. Eveleigh, Mouse ENU Mutagenesis to Understand Immunity to Infection: Methods, Selected Examples, and Perspectives, Genes, vol.5, pp.887-925, 2014.

G. Caignard, G. A. Leiva-torres, M. Leney-greene, and B. Charbonneau, Genome-wide mouse mutagenesis reveals CD45-mediated T cell function as critical in protective immunity to HSV-1, PLoS pathogens, vol.9, p.1003637, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-01130346

L. C. Caires-junior, E. Goulart, U. S. Melo, and B. Araujo, Discordant congenital Zika syndrome twins show differential in vitro viral susceptibility of neural progenitor cells, Nature communications, vol.9, p.475, 2018.

G. S. Campos, A. C. Bandeira, and S. I. Sardi, Zika Virus Outbreak, vol.21, pp.1885-1886, 2015.

G. Canguilhem, Etudes d'histoire et de philosophie des sciences concernant les vivants et la vie, 1968.

V. M. Cao-lormeau, A. Blake, S. Mons, and S. Lastere, Guillain-Barre Syndrome outbreak associated with Zika virus infection in French Polynesia: a case-control study, Lancet, vol.387, pp.1531-1539, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01453407

V. M. Cao-lormeau, C. Roche, A. Teissier, and E. Robin, Zika virus, French polynesia, South pacific, Emerging infectious diseases, vol.20, pp.1085-1086, 2013.

B. Cao, L. A. Parnell, M. S. Diamond, and I. U. Mysorekar, Inhibition of autophagy limits vertical transmission of Zika virus in pregnant mice, The Journal of experimental medicine, vol.214, pp.2303-2313, 2017.

J. L. Casanova, L. Abel, and L. Quintana-murci, Immunology taught by human genetics, Cold Spring Harbor symposia on quantitative biology, vol.78, pp.157-172, 2013.

D. D. Cavalcanti, L. V. Alves, G. J. Furtado, and C. C. Santos, Echocardiographic findings in infants with presumed congenital Zika syndrome: Retrospective case series study, PLoS One, vol.12, p.175065, 2017.

M. G. Cavalcanti, M. J. Cabral-castro, J. Goncalves, L. S. Santana, E. S. Pimenta et al., Zika virus shedding in human milk during lactation: an unlikely source of infection?, International journal of infectious diseases : IJID : official publication of the International Society for Infectious Diseases, vol.57, pp.70-72, 2017.

, Emerging Infectious Diseases

J. Cerbino-neto, E. C. Mesquita, T. M. Souza, and V. Parreira, Clinical Manifestations of Zika Virus Infection, Emerging infectious diseases, vol.22, pp.1318-1320, 2015.

J. F. Chan, A. J. Zhang, C. C. Chan, and C. C. Yip, Zika Virus Infection in Dexamethasoneimmunosuppressed Mice Demonstrating Disseminated Infection with Multi-organ Involvement Including Orchitis Effectively Treated by Recombinant Type I Interferons, EBioMedicine, vol.14, pp.112-122, 2016.

S. J. Chapman and A. V. Hill, Human genetic susceptibility to infectious disease, Nat Rev Genet, vol.13, pp.175-188, 2012.

M. Chemudupati, A. D. Kenney, S. Bonifati, A. Zani, T. M. Mcmichael et al., From APOBEC to ZAP: Diverse mechanisms used by cellular restriction factors to inhibit virus infections. Biochimica et biophysica acta, Molecular cell research, vol.1866, pp.382-394, 2019.

E. J. Chesler, D. R. Miller, L. R. Branstetter, and L. D. Galloway, The Collaborative Cross at Oak Ridge National Laboratory: developing a powerful resource for systems genetics, Mamm Genome, vol.19, pp.382-389, 2008.

L. Chevallier, C. Blanchet, J. Jaubert, and E. Pachulec, Resistance to plague of Mus spretus SEG/Pas mice requires the combined action of at least four genetic factors, Genes and immunity, vol.14, pp.35-41, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-02073274

J. P. Chippaux and A. Chippaux, Yellow fever in Africa and the Americas: a historical and epidemiological perspective. The journal of venomous animals and toxins including tropical diseases, vol.24, p.20, 2018.

G. A. Churchill, D. C. Airey, H. Allayee, and J. M. Angel, The Collaborative Cross, a community resource for the genetic analysis of complex traits, Nature genetics, vol.36, pp.1133-1137, 2004.

M. J. Ciancanelli, L. Abel, S. Y. Zhang, and J. L. Casanova, Host genetics of severe influenza: from mouse Mx1 to human IRF7, Curr Opin Immunol, vol.38, pp.109-120, 2016.

M. J. Ciancanelli, S. X. Huang, P. Luthra, and H. Garner, Infectious disease. Life-threatening influenza and impaired interferon amplification in human IRF7 deficiency, Science, vol.348, pp.448-453, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02517129

M. Civelek and A. J. Lusis, Systems genetics approaches to understand complex traits, Nat Rev Genet, vol.15, pp.34-48, 2014.

C. S. Clancy, A. J. Van-wettere, J. D. Morrey, and J. G. Julander, Coitus-Free Sexual Transmission of Zika Virus in a Mouse Model, Scientific reports, vol.8, p.15379, 2018.

C. S. Clancy, A. J. Van-wettere, J. D. Morrey, and J. G. Julander, Zika Virus Associated Pathology and Antigen Presence in the Testicle in the Absence of Sexual Transmission During Subacute to Chronic Infection in a Mouse Model, Scientific reports, vol.9, p.8325, 2019.

M. Clementi, D. Gianantonio, and E. , Genetic susceptibility to infectious diseases, Reproductive toxicology, vol.21, pp.345-349, 2006.

L. L. Coffey, R. I. Keesler, P. A. Pesavento, and K. Woolard, Intraamniotic Zika virus inoculation of pregnant rhesus macaques produces fetal neurologic disease, Nature communications, vol.9, p.2414, 2018.

C. Cross-consortium, The genome architecture of the Collaborative Cross mouse genetic reference population, Genetics, vol.190, pp.389-401, 2012.

R. Collin, L. Balmer, G. Morahan, and S. Lesage, Common Heritable Immunological Variations Revealed in Genetically Diverse Inbred Mouse Strains of the Collaborative Cross, Journal of immunology, vol.202, pp.777-786, 1950.

D. Concepcion, K. L. Seburn, G. Wen, W. N. Frankel, and B. A. Hamilton, Mutation rate and predicted phenotypic target sizes in ethylnitrosourea-treated mice, Genetics, vol.168, pp.953-959, 2004.

B. Connor and W. B. Bunn, The changing epidemiology of Japanese encephalitis and New data: the implications for New recommendations for Japanese encephalitis vaccine. Tropical diseases, vol.3, p.14, 2017.

C. B. Coyne and H. M. Lazear, Zika virus -reigniting the TORCH, Nat Rev Microbiol, vol.14, pp.707-715, 2016.

Y. J. Crow and N. Manel, Aicardi-Goutieres syndrome and the type I interferonopathies, Nat Rev Immunol, vol.15, pp.429-440, 2015.

F. R. Cugola, I. R. Fernandes, F. B. Russo, and B. C. Freitas, The Brazilian Zika virus strain causes birth defects in experimental models, Nature, vol.534, pp.267-271, 2016.

L. Cui, P. Zou, E. Chen, and H. Yao, Visual and Motor Deficits in Grown-up Mice with Congenital Zika Virus Infection, EBioMedicine, vol.20, pp.193-201, 2017.

S. L. Cumberworth, J. J. Clark, A. Kohl, and C. L. Donald, Inhibition of type I interferon induction and signalling by mosquito-borne flaviviruses, Cellular microbiology, p.19, 2017.

I. Da-silva, J. A. Frontera, A. M. Bispo-de-filippis, and O. Nascimento, Neurologic Complications Associated With the Zika Virus in Brazilian Adults, JAMA neurology, vol.74, pp.1190-1198, 2017.

J. Dang, S. K. Tiwari, G. Lichinchi, Y. Qin, V. S. Patil et al., Zika Virus Depletes Neural Progenitors in Human Cerebral Organoids through Activation of the Innate Immune Receptor TLR3, Cell stem cell, vol.19, pp.258-265, 2016.

J. Darbellay, B. Cox, K. Lai, and M. Delgado-ortega, Zika Virus Causes Persistent Infection in Porcine Conceptuses and may Impair Health in Offspring, EBioMedicine, vol.25, pp.73-86, 2017.

S. David and A. M. Abraham, Epidemiological and clinical aspects on West Nile virus, a globally emerging pathogen, vol.48, pp.571-586, 2016.

W. K. De-oliveira, G. De-franca, E. H. Carmo, B. B. Duncan, R. De-souza-kuchenbecker et al., Infection-related microcephaly after the 2015 and 2016 Zika virus outbreaks in Brazil: a surveillance-based analysis, Lancet, vol.390, pp.861-870, 2017.

M. J. De-veer, M. Holko, M. Frevel, and E. Walker, Functional classification of interferonstimulated genes identified using microarrays, Journal of leukocyte biology, vol.69, pp.912-920, 2001.

M. Dean, M. Carrington, C. Winkler, and G. A. Huttley, Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia Growth and Development Study, Multicenter AIDS Cohort Study, ALIVE Study. Science, vol.273, pp.1856-1862, 1996.

D. Campo, M. Feitosa, I. M. Ribeiro, E. M. Horovitz, and D. D. , The phenotypic spectrum of congenital Zika syndrome, American journal of medical genetics. Part A, vol.173, pp.841-857, 2017.

E. Delatorre, D. Mir, and G. Bello, Tracing the origin of the NS1 A188V substitution responsible for recent enhancement of Zika virus Asian genotype infectivity, Memorias do Instituto Oswaldo Cruz, vol.112, pp.793-795, 2017.

D. Santo, J. P. Apetrei, and C. , Animal models for viral diseases: Non-human primate and humanized mouse models for viral infections, Curr Opin Virol, vol.25, 2017.

G. W. Dick, S. F. Kitchen, and A. J. Haddow, Zika virus. I. Isolations and serological specificity, Transactions of the Royal Society of Tropical Medicine and Hygiene, vol.46, pp.509-520, 1952.

Q. Ding, J. M. Gaska, F. Douam, and L. Wei, Species-specific disruption of STING-dependent antiviral cellular defenses by the Zika virus NS2B3 protease, Proceedings of the National Academy of Sciences of the United States of America, vol.115, pp.6310-6318, 2018.

E. Dirlikov, C. G. Major, N. A. Medina, and R. Lugo-robles, Clinical Features of Guillain-Barre Syndrome With vs Without Zika Virus Infection, JAMA neurology, vol.75, pp.1089-1097, 2016.

C. L. Donald, B. Brennan, S. L. Cumberworth, and V. V. Rezelj, Full Genome Sequence and sfRNA Interferon Antagonist Activity of Zika Virus from Recife, Brazil, PLoS neglected tropical diseases, vol.10, p.5048, 2016.

D. Santos, T. Rodriguez, A. Almiron, M. Sanhueza, and A. , Zika Virus and the Guillain-Barre Syndrome -Case Series from Seven Countries, N Engl J Med, vol.375, pp.1598-1601, 2016.

S. D. Dowall, V. A. Graham, E. Rayner, and B. Atkinson, A Susceptible Mouse Model for Zika Virus Infection, PLoS neglected tropical diseases, vol.10, p.4658, 2016.

S. D. Dowall, V. A. Graham, E. Rayner, and L. Hunter, Lineage-dependent differences in the disease progression of Zika virus infection in type-I interferon receptor knockout (A129) mice, PLoS neglected tropical diseases, vol.11, p.5704, 2017.

R. W. Driggers, C. Y. Ho, E. M. Korhonen, and S. Kuivanen, Zika Virus Infection with Prolonged Maternal Viremia and Fetal Brain Abnormalities, N Engl J Med, vol.374, pp.2142-2151, 2016.

S. B. Drutman, F. Haerynck, F. L. Zhong, and D. Hum, Homozygous NLRP1 gain-of-function mutation in siblings with a syndromic form of recurrent respiratory papillomatosis, Proceedings of the National Academy of Sciences of the United States of America, vol.116, pp.19055-19063, 2019.

D. M. Dudley, M. T. Aliota, E. L. Mohr, C. M. Newman, T. G. Golos et al., , 2019.

, Using Macaques to Address Critical Questions in Zika Virus Research, Annual review of virology, vol.6, pp.481-500

D. M. Dudley, M. T. Aliota, E. L. Mohr, and A. M. Weiler, A rhesus macaque model of Asianlineage Zika virus infection, Nature communications, vol.7, p.12204, 2016.

M. R. Duffy, T. H. Chen, W. T. Hancock, and A. M. Powers, Zika virus outbreak on Yap Island, Federated States of Micronesia, N Engl J Med, vol.360, pp.2536-2543, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-00734543

J. H. Dufour, M. Dziejman, M. T. Liu, J. H. Leung, T. E. Lane et al., , 2002.

, CXCL10)-deficient mice reveal a role for IP-10 in effector T cell generation and trafficking, Journal of immunology, vol.168, pp.3195-3204, 1950.

N. K. Duggal, E. M. Mcdonald, J. M. Ritter, and A. C. Brault, Sexual transmission of Zika virus enhances in utero transmission in a mouse model, Scientific reports, vol.8, p.4510, 2018.

M. Dupont-rouzeyrol, A. Biron, O. O'connor, E. Huguon, and E. Descloux, Infectious Zika viral particles in breastmilk, Lancet, vol.387, p.1051, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01634326

M. Dupont-rouzeyrol, O. Connor, O. Calvez, E. Daures, M. John et al., Co-infection with Zika and dengue viruses in 2 patients, Emerging infectious diseases, vol.21, pp.381-382, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01114780

C. Durrant, H. Tayem, B. Yalcin, and J. Cleak, Collaborative Cross mice and their power to map host susceptibility to Aspergillus fumigatus infection, Genome Res, vol.21, pp.1239-1248, 2011.

G. Eberl, M. Colonna, D. Santo, J. P. Mckenzie, and A. N. , Innate lymphoid cells. Innate lymphoid cells: a new paradigm in immunology, Science, vol.348, p.6566, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-02101002

H. M. El-sahly, R. Gorchakov, L. Lai, and M. S. Natrajan, Clinical, Virologic, and Immunologic Characteristics of Zika Virus Infection in a Cohort of US Patients: Prolonged RNA Detection in Whole Blood, Open forum infectious diseases, vol.6, p.352, 2019.

H. Elbahesh and K. Schughart, Genetically diverse CC-founder mouse strains replicate the human influenza gene expression signature, Scientific reports, vol.6, p.26437, 2016.

E. Ngono, A. Vizcarra, E. A. Tang, W. W. Sheets, and N. , Mapping and Role of the CD8+ T Cell Response During Primary Zika Virus Infection in Mice, Cell host & microbe, vol.21, pp.35-46, 2017.

J. Ermann and L. H. Glimcher, After GWAS: mice to the rescue?, Curr Opin Immunol, vol.24, pp.564-570, 2012.

S. Eskildsen, J. Justesen, M. H. Schierup, and R. Hartmann, Characterization of the 2'-5'-oligoadenylate synthetase ubiquitin-like family, Nucleic acids research, vol.31, pp.3166-3173, 2003.

, Press Release, 2019.

A. R. Everitt, C. S. Pertel, T. John, and S. P. , IFITM3 restricts the morbidity and mortality associated with influenza, Nature, vol.484, pp.519-523, 2012.

O. Faye, C. C. Freire, A. Iamarino, and O. Faye, Molecular evolution of Zika virus during its emergence in the 20(th) century, PLoS neglected tropical diseases, vol.8, p.2636, 2014.

J. Fellay, K. V. Shianna, D. Ge, and S. Colombo, A whole-genome association study of major determinants for host control of HIV-1, Science, vol.317, pp.944-947, 2007.

V. Fensterl, S. Chattopadhyay, and G. C. Sen, No Love Lost Between Viruses and Interferons, Annual review of virology, vol.2, pp.549-572, 2015.

N. C. Fernandes, J. S. Nogueira, R. A. Ressio, and C. S. Cirqueira, Experimental Zika virus infection induces spinal cord injury and encephalitis in newborn Swiss mice, Experimental and toxicologic pathology : official journal of the Gesellschaft fur Toxikologische Pathologie, vol.69, pp.63-71, 2017.

A. C. Ferreira, C. Zaverucha-do-valle, P. A. Reis, and G. Barbosa-lima, Sofosbuvir protects Zika virus-infected mice from mortality, preventing short-and long-term sequelae, Scientific reports, vol.7, p.9409, 2017.

M. T. Ferris, D. L. Aylor, D. Bottomly, and A. C. Whitmore, Modeling host genetic regulation of influenza pathogenesis in the collaborative cross, PLoS pathogens, vol.9, p.1003196, 2013.

C. Flamand, C. Fritzell, S. Matheus, and M. Dueymes, The proportion of asymptomatic infections and spectrum of disease among pregnant women infected by Zika virus: systematic monitoring in French Guiana, Euro surveillance : bulletin Europeen sur les maladies transmissibles = European communicable disease bulletin, p.22, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02539219

J. Flint and E. Eskin, Genome-wide association studies in mice, Nat Rev Genet, vol.13, pp.807-817, 2012.

T. Fourie, G. Grard, I. Leparc-goffart, S. Briolant, and A. Fontaine, Variability of Zika Virus Incubation Period in Humans, Open forum infectious diseases, vol.5, p.261, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02374799

B. D. Foy, K. C. Kobylinski, C. Foy, J. L. Blitvich, and B. J. , Probable non-vector-borne transmission of Zika virus, Emerging infectious diseases, vol.17, pp.880-882, 2011.

J. E. French, D. M. Gatti, D. L. Morgan, and G. E. Kissling, Diversity Outbred Mice Identify Population-Based Exposure Thresholds and Genetic Factors that Influence Benzene-Induced Genotoxicity, Environmental health perspectives, vol.123, pp.237-245, 2015.

J. M. Furtado, D. L. Esposito, T. M. Klein, T. Teixeira-pinto, and B. A. Da-fonseca, Uveitis Associated with Zika Virus Infection, N Engl J Med, vol.375, pp.394-396, 2016.

M. U. Gack and M. S. Diamond, Innate immune escape by Dengue and West Nile viruses, Curr Opin Virol, vol.20, pp.119-128, 2016.

C. Garber, A. Soung, L. L. Vollmer, M. Kanmogne, A. Last et al., T cells promote microglia-mediated synaptic elimination and cognitive dysfunction during recovery from neuropathogenic flaviviruses, Nature neuroscience, vol.22, pp.1276-1288, 2019.

P. P. Garcez, E. C. Loiola, M. Da-costa, R. Higa, and L. M. , Zika virus impairs growth in human neurospheres and brain organoids, vol.352, pp.816-818, 2016.

M. R. Gaudinski, K. V. Houser, K. M. Morabito, and Z. Hu, Safety, tolerability, and immunogenicity of two Zika virus DNA vaccine candidates in healthy adults: randomised, open-label, phase 1 clinical trials, Lancet, vol.391, pp.552-562, 2018.

M. Giovanetti, G. De-jesus, J. , L. De-maia, M. Junior et al., Genetic evidence of Zika virus in mother's breast milk and body fluids of a newborn with severe congenital defects. Clinical microbiology and infection : the official publication of the, European Society of Clinical Microbiology and Infectious Diseases, vol.24, pp.1111-1112, 2018.

I. Gladwyn-ng, L. Cordon-barris, C. Alfano, and C. Creppe, Stress-induced unfolded protein response contributes to Zika virus-associated microcephaly, Nature neuroscience, vol.21, pp.63-71, 2018.
URL : https://hal.archives-ouvertes.fr/pasteur-01741181

S. Glineur, N. Antoine-moussiaux, C. Michaux, and D. Desmecht, Immune depression of the SJL/J mouse, a radioresistant and immunologically atypical inbred strain, Immunobiology, vol.216, pp.213-217, 2011.

A. P. Goncalvez, R. E. Engle, M. St-claire, R. H. Purcell, and C. J. Lai, Monoclonal antibody-mediated enhancement of dengue virus infection in vitro and in vivo and strategies for prevention, Proceedings of the National Academy of Sciences of the United States of America, vol.104, pp.9422-9427, 2007.

A. B. Goodman, E. J. Dziuban, K. Powell, and R. H. Bitsko, Characteristics of Children Aged <18 Years with Zika Virus Disease Acquired Postnatally -U.S. States, MMWR. Morbidity and mortality weekly report, vol.65, pp.1082-1085, 2015.

M. J. Gorman, E. A. Caine, K. Zaitsev, and M. C. Begley, An Immunocompetent Mouse Model of Zika Virus Infection, Cell host & microbe, vol.23, pp.672-685, 2018.

J. Govero, P. Esakky, S. M. Scheaffer, and E. Fernandez, Zika virus infection damages the testes in mice, Nature, vol.540, pp.438-442, 2016.

J. B. Graham, J. L. Swarts, M. Mooney, and G. Choonoo, Extensive Homeostatic T Cell Phenotypic Variation within the Collaborative Cross, Cell reports, vol.21, pp.2313-2325, 2017.

J. B. Graham, J. L. Swarts, C. Wilkins, and S. Thomas, A Mouse Model of Chronic West Nile Virus Disease, PLoS pathogens, vol.12, p.1005996, 2016.

J. B. Graham, S. Thomas, J. Swarts, and A. A. Mcmillan, Genetic diversity in the collaborative cross model recapitulates human West Nile virus disease outcomes, vol.6, pp.493-00415, 2015.

L. E. Gralinski, M. T. Ferris, D. L. Aylor, and A. C. Whitmore, Genome Wide Identification of SARS-CoV Susceptibility Loci Using the Collaborative Cross, PLoS genetics, vol.11, p.1005504, 2015.

A. Grant, S. S. Ponia, S. Tripathi, and V. Balasubramaniam, Zika Virus Targets Human STAT2 to Inhibit Type I Interferon Signaling, Cell host & microbe, vol.19, pp.882-890, 2016.

R. Greek, Animal Models in Drug Development, 2013.

R. Green, R. C. Ireton, M. Gale, and J. , Interferon-stimulated genes: new platforms and computational approaches, Mamm Genome, vol.29, pp.593-602, 2018.

R. Green, C. Wilkins, S. Thomas, and A. Sekine, Oas1b-dependent Immune Transcriptional Profiles of West Nile Virus Infection in the Collaborative Cross, G3 (Bethesda), vol.7, pp.1665-1682, 2017.

A. Grifoni, J. Pham, J. Sidney, and P. H. O&apos;rourke, Prior Dengue Virus Exposure Shapes T Cell Immunity to Zika Virus in Humans, Journal of virology, p.91, 2017.

S. Group, Outbreak of Zika virus infection in Singapore: an epidemiological, entomological, virological, and clinical analysis, The Lancet. Infectious diseases, vol.17, pp.813-821, 2017.

N. D. Grubaugh, J. T. Ladner, M. Kraemer, and G. Dudas, Genomic epidemiology reveals multiple introductions of Zika virus into the United States, Nature, vol.546, pp.401-405, 2017.

S. Gurung, N. Reuter, A. Preno, and J. Dubaut, Zika virus infection at mid-gestation results in fetal cerebral cortical injury and fetal death in the olive baboon, PLoS pathogens, vol.15, p.1007507, 2019.

G. Gutierrez-bugallo, L. A. Piedra, M. Rodriguez, and J. A. Bisset, Vector-borne transmission and evolution of Zika virus, Nature ecology & evolution, vol.3, pp.561-569, 2019.

D. G. Hackam and D. A. Redelmeier, Translation of research evidence from animals to humans, Jama, vol.296, pp.1731-1732, 2006.

A. D. Haddow, A. J. Schuh, C. Y. Yasuda, and M. R. Kasper, Genetic characterization of Zika virus strains: geographic expansion of the Asian lineage, PLoS neglected tropical diseases, vol.6, p.1477, 2012.

U. A. Halai, K. Nielsen-saines, M. L. Moreira, and P. C. De-sequeira, Maternal Zika Virus Disease Severity, Virus Load, Prior Dengue Antibodies, and Their Relationship to Birth Outcomes, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America, vol.65, pp.877-883, 2017.

M. Harries and I. Smith, The development and clinical use of trastuzumab (Herceptin), Endocrinerelated cancer, vol.9, pp.75-85, 2002.

S. S. Hasan, M. Sevvana, R. J. Kuhn, and M. G. Rossmann, Structural biology of Zika virus and other flaviviruses, Nature structural & molecular biology, vol.25, pp.13-20, 2018.

V. Heang, C. Y. Yasuda, L. Sovann, A. D. Haddow, T. Da-rosa et al., Emerging infectious diseases, vol.18, pp.349-351, 2010.

B. A. Hendrickson, J. Guo, I. Brown, K. Dennis, D. Marcellino et al., Decreased vaginal disease in J-chain-deficient mice following herpes simplex type 2 genital infection, Virology, vol.271, pp.155-162, 2000.

M. Hennessey, M. Fischer, and J. E. Staples, Zika Virus Spreads to New Areas -Region of the Americas, vol.65, pp.55-58, 2015.

T. P. Hill, Phase 0 clinical trials: towards a more complete ethics critique, vol.6, p.248, 2012.

A. J. Hirsch, V. Roberts, P. L. Grigsby, and N. Haese, Zika virus infection in pregnant rhesus macaques causes placental dysfunction and immunopathology, Nature communications, vol.9, p.263, 2018.

M. R. Hobbs, B. B. Jones, B. E. Otterud, M. Leppert, and J. D. Kriesel, Identification of a herpes simplex labialis susceptibility region on human chromosome 21, The Journal of infectious diseases, vol.197, pp.340-346, 2008.

B. Hoen, B. Schaub, A. L. Funk, and V. Ardillon, Pregnancy Outcomes after ZIKV Infection in French Territories in the Americas, N Engl J Med, vol.378, pp.985-994, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01744446

M. A. Honein, A. L. Dawson, E. E. Petersen, and A. M. Jones, Birth Defects Among Fetuses and Infants of US Women With Evidence of Possible Zika Virus Infection During Pregnancy, Jama, vol.317, pp.59-68, 2017.

M. A. Horisberger, P. Staeheli, and O. Haller, Interferon induces a unique protein in mouse cells bearing a gene for resistance to influenza virus, Proceedings of the National Academy of Sciences of the United States of America, vol.80, pp.1910-1914, 1983.

H. Huang, S. Li, Y. Zhang, and X. Han, CD8(+) T Cell Immune Response in Immunocompetent Mice during Zika Virus Infection, Journal of virology, p.91, 2017.

W. C. Huang, R. Abraham, B. S. Shim, H. Choe, and D. T. Page, Zika virus infection during the period of maximal brain growth causes microcephaly and corticospinal neuron apoptosis in wild type mice, Scientific reports, vol.6, p.34793, 2016.

Y. S. Huang, S. Higgs, and D. L. Vanlandingham, Emergence and re-emergence of mosquito-borne arboviruses, Curr Opin Virol, vol.34, pp.104-109, 2019.

N. P. Hudson, The Pathology of Experimental Yellow Fever in the Macacus Rhesus: I. Gross Pathology, The American journal of pathology, vol.4, pp.395-406, 1928.

G. D. Huhn, J. J. Sejvar, S. P. Montgomery, and M. S. Dworkin, West Nile virus in the United States: an update on an emerging infectious disease, American family physician, vol.68, pp.653-660, 2003.

P. R. Hutchings, A. M. Varey, and A. Cooke, Immunological defects in SJL mice, Immunology, vol.59, pp.445-450, 1986.

J. Ikejezie, C. N. Shapiro, J. Kim, and M. Chiu, Zika Virus Transmission -Region of the Americas, MMWR. Morbidity and mortality weekly report, vol.66, pp.329-334, 2015.

F. A. Iraqi, G. Churchill, and R. Mott, The Collaborative Cross, developing a resource for mammalian systems genetics: a status report of the Wellcome Trust cohort, Mamm Genome, vol.19, pp.379-381, 2008.

A. S. Jaeger, R. A. Murrieta, L. R. Goren, and C. M. Crooks, Zika viruses of African and Asian lineages cause fetal harm in a mouse model of vertical transmission, PLoS neglected tropical diseases, vol.13, p.7343, 2019.

B. W. Jagger, J. J. Miner, B. Cao, and N. Arora, Gestational Stage and IFN-lambda Signaling Regulate ZIKV Infection In Utero, Cell host & microbe, vol.22, pp.366-376, 2017.

E. S. Jentes, G. Poumerol, M. D. Gershman, and D. R. Hill, The revised global yellow fever risk map and recommendations for vaccination, 2010: consensus of the Informal WHO Working Group on Geographic Risk for Yellow Fever, The Lancet. Infectious diseases, vol.11, pp.622-632, 2011.

K. A. Jurado, L. J. Yockey, P. W. Wong, S. Lee, A. J. Huttner et al., Antiviral CD8 T cells induce Zika-virus-associated paralysis in mice, Nature microbiology, vol.3, pp.141-147, 2018.

Y. W. Kam, C. Y. Lee, T. H. Teo, and S. W. Howland, Cross-reactive dengue human monoclonal antibody prevents severe pathologies and death from Zika virus infections, JCI insight, vol.2, 2017.

N. Kamiyama, R. Soma, S. Hidano, and K. Watanabe, Ribavirin inhibits Zika virus (ZIKV) replication in vitro and suppresses viremia in ZIKV-infected STAT1-deficient mice, Antiviral Res, vol.146, pp.1-11, 2017.

D. T. Karamitsos, The story of insulin discovery, Diabetes research and clinical practice, vol.93, pp.2-8, 2011.

O. Karimi, A. Goorhuis, J. Schinkel, and J. Codrington, Thrombocytopenia and subcutaneous bleedings in a patient with Zika virus infection, Lancet, vol.387, pp.939-940, 2016.

M. P. Karwowski, J. M. Nelson, J. E. Staples, and M. Fischer, Zika Virus Disease: A CDC Update for Pediatric Health Care Providers, Pediatrics, vol.137, 2016.

H. Kato, O. Takeuchi, S. Sato, and M. Yoneyama, Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses, Nature, vol.441, pp.101-105, 2006.

L. C. Katzelnick, L. Gresh, M. E. Halloran, and J. C. Mercado, Antibody-dependent enhancement of severe dengue disease in humans, Science, vol.358, pp.929-932, 2017.

E. B. Kauffman and L. D. Kramer, Zika Virus Mosquito Vectors: Competence, Biology, and Vector Control, The Journal of infectious diseases, vol.216, pp.976-990, 2017.

A. B. Kawiecki, E. H. Mayton, M. F. Dutuze, B. A. Goupil, I. M. Langohr et al., Tissue tropisms, infection kinetics, histologic lesions, and antibody response of the MR766 strain of Zika virus in a murine model, Virology journal, vol.14, p.82, 2017.

G. R. Keele, W. L. Crouse, S. Kelada, and W. Valdar, Determinants of QTL Mapping Power in the Realized Collaborative Cross, Bethesda), vol.3, pp.1707-1727, 2019.

A. D. Kenney, J. A. Dowdle, L. Bozzacco, and T. M. Mcmichael, Human Genetic Determinants of Viral Diseases, Annual review of genetics, vol.51, pp.241-263, 2017.

S. Khan, E. M. Woodruff, M. Trapecar, and K. A. Fontaine, Dampened antiviral immunity to intravaginal exposure to RNA viral pathogens allows enhanced viral replication, The Journal of experimental medicine, vol.213, pp.2913-2929, 2016.

S. F. Kluge, D. Sauter, and F. Kirchhoff, SnapShot: antiviral restriction factors, Cell, vol.163, pp.774-774, 2015.

S. Kodati, T. N. Palmore, F. A. Spellman, D. Cunningham, B. Weistrop et al., Bilateral posterior uveitis associated with Zika virus infection, Lancet, vol.389, pp.125-126, 2017.

F. Koide, S. Goebel, B. Snyder, and K. B. Walters, Development of a Zika Virus Infection Model in Cynomolgus Macaques, Front Microbiol, vol.7, p.2028, 2016.

H. Kollmus, C. Pilzner, S. R. Leist, M. Heise, R. Geffers et al., Of mice and men: the host response to influenza virus infection, Mamm Genome, vol.29, pp.446-470, 2018.

M. Kraemer, R. C. Reiner, J. Brady, O. J. Messina, and J. P. , Past and future spread of the arbovirus vectors Aedes aegypti and Aedes albopictus, Nature microbiology, vol.4, pp.854-863, 2019.
URL : https://hal.archives-ouvertes.fr/pasteur-02067318

A. Krishnankutty, T. Kimura, T. Saito, and K. Aoyagi, In vivo regulation of glycogen synthase kinase 3beta activity in neurons and brains, Scientific reports, vol.7, p.8602, 2017.

J. Kristic, O. O. Zaytseva, R. Ram, and Q. Nguyen, Profiling and genetic control of the murine immunoglobulin G glycome, Nature chemical biology, vol.14, pp.516-524, 2018.

A. Kumar, S. Hou, A. M. Airo, and D. Limonta, Zika virus inhibits type-I interferon production and downstream signaling, EMBO reports, vol.17, pp.1766-1775, 2016.

M. Kumar, K. K. Krause, F. Azouz, E. Nakano, and V. R. Nerurkar, A guinea pig model of Zika virus infection, Virology journal, vol.14, p.75, 2017.

R. S. Lanciotti, O. L. Kosoy, J. J. Laven, and J. O. Velez, Genetic and serologic properties of Zika virus associated with an epidemic, Yap State, Micronesia, Emerging infectious diseases, vol.14, pp.1232-1239, 2007.

T. Langerak, N. Mumtaz, V. I. Tolk, E. Van-gorp, B. E. Martina et al., The possible role of cross-reactive dengue virus antibodies in Zika virus pathogenesis, PLoS pathogens, vol.15, p.1007640, 2019.

A. Lannuzel, J. L. Ferge, Q. Lobjois, and A. Signate, Long-term outcome in neuroZika: When biological diagnosis matters, Neurology, vol.92, pp.2406-2420, 2019.
URL : https://hal.archives-ouvertes.fr/inserm-02338513

R. A. Larocca, P. Abbink, J. P. Peron, and P. M. Zanotto, Vaccine protection against Zika virus from Brazil, Nature, vol.536, pp.474-478, 2016.

M. Laureti, D. Narayanan, J. Rodriguez-andres, J. K. Fazakerley, and L. Kedzierski, Flavivirus Receptors: Diversity, Identity, and Cell Entry, vol.9, p.2180, 2018.

H. M. Lazear, J. Govero, A. M. Smith, D. J. Platt, E. Fernandez et al., A Mouse Model of Zika Virus Pathogenesis, Cell host & microbe, vol.19, pp.720-730, 2016.

L. Flohic, G. Porphyre, V. Barbazan, P. Gonzalez, and J. P. , Review of climate, landscape, and viral genetics as drivers of the Japanese encephalitis virus ecology, PLoS neglected tropical diseases, vol.7, p.2208, 2013.

S. R. Leist and R. S. Baric, Giving the Genes a Shuffle: Using Natural Variation to Understand Host Genetic Contributions to Viral Infections, Trends in genetics : TIG, vol.34, pp.777-789, 2018.

S. R. Leist, C. Pilzner, J. M. Van-den-brand, and L. Dengler, Influenza H3N2 infection of the collaborative cross founder strains reveals highly divergent host responses and identifies a unique phenotype in CAST/EiJ mice, BMC genomics, vol.17, p.143, 2016.

C. Li, Y. Q. Deng, S. Wang, and F. Ma, 25-Hydroxycholesterol Protects Host against Zika Virus Infection and Its Associated Microcephaly in a Mouse Model, Immunity, vol.46, pp.446-456, 2017.

C. Li, Q. Wang, Y. Jiang, and Q. Ye, Disruption of glial cell development by Zika virus contributes to severe microcephalic newborn mice, Cell discovery, vol.4, p.43, 2018.

C. Li, D. Xu, Q. Ye, and S. Hong, Zika Virus Disrupts Neural Progenitor Development and Leads to Microcephaly in Mice, Cell stem cell, vol.19, pp.120-126, 2016.

H. Li, L. Saucedo-cuevas, J. A. Regla-nava, and G. Chai, Zika Virus Infects Neural Progenitors in the Adult Mouse Brain and Alters Proliferation, Cell stem cell, vol.19, pp.593-598, 2016.

S. Li, N. Armstrong, H. Zhao, and W. Hou, Zika Virus Fatally Infects Wild Type Neonatal Mice and Replicates in Central Nervous System, Viruses, vol.10, 2018.

X. F. Li, H. L. Dong, X. Y. Huang, and Y. F. Qiu, Characterization of a 2016 Clinical Isolate of Zika Virus in Non-human, Primates. EBioMedicine, vol.12, pp.170-177, 2016.

J. Lilue, A. G. Doran, I. T. Fiddes, and M. Abrudan, Sixteen diverse laboratory mouse reference genomes define strain-specific haplotypes and novel functional loci, Nature genetics, vol.50, pp.1574-1583, 2018.

J. Lindenmann, C. A. Lane, and D. Hobson, THE RESISTANCE OF A2G MICE TO MYXOVIRUSES, Journal of immunology, vol.90, pp.942-951, 1950.

R. Lindqvist, C. Kurhade, J. D. Gilthorpe, and A. K. Overby, Cell-type-and region-specific restriction of neurotropic flavivirus infection by viperin, Journal of neuroinflammation, vol.15, p.80, 2018.

R. Lindqvist, F. Mundt, J. D. Gilthorpe, S. Wolfel, N. O. Gekara et al., Fast type I interferon response protects astrocytes from flavivirus infection and virus-induced cytopathic effects, Journal of neuroinflammation, vol.13, p.277, 2016.

Y. Liu, J. Liu, S. Du, and C. Shan, Evolutionary enhancement of Zika virus infectivity in Aedes aegypti mosquitoes, Nature, vol.545, pp.482-486, 2017.

Z. Y. Liu, W. F. Shi, and C. F. Qin, The evolution of Zika virus from Asia to the Americas, Nat Rev Microbiol, vol.17, pp.131-139, 2019.

L. A. Low and D. A. Tagle, Organs-on-Chips") for Drug Efficacy and Toxicity Testing, Clinical and translational science, vol.10, pp.237-239, 2017.

J. Ma, H. Ketkar, T. Geng, and E. Lo, Zika Virus Non-structural Protein 4A Blocks the RLR-MAVS Signaling, Front Microbiol, vol.9, p.1350, 2018.

W. Ma, S. Li, S. Ma, and L. Jia, Zika Virus Causes Testis Damage and Leads to Male Infertility in Mice, Cell, vol.167, pp.1511-1524, 1510.

F. N. Macnamara, Zika virus: a report on three cases of human infection during an epidemic of jaundice in Nigeria, Transactions of the Royal Society of Tropical Medicine and Hygiene, vol.48, pp.139-145, 1954.

H. Malkki, CNS infections: Mouse studies confirm the link between Zika virus infection and microcephaly, Nat Rev Neurol, vol.12, p.369, 2016.

M. Manangeeswaran, D. D. Ireland, and D. Verthelyi, Zika (PRVABC59) Infection Is Associated with T cell Infiltration and Neurodegeneration in CNS of Immunocompetent Neonatal C57Bl/6 Mice, PLoS pathogens, vol.12, p.1006004, 2016.

C. Manet, C. Roth, A. Tawfik, T. Cantaert, A. Sakuntabhai et al., Host genetic control of mosquito-borne Flavivirus infections, Mamm Genome, vol.29, pp.384-407, 2018.
URL : https://hal.archives-ouvertes.fr/pasteur-02003816

C. Manet, E. Simon-loriere, G. Jouvion, and D. Hardy, Genetic diversity of Collaborative Cross mice controls viral replication, clinical severity and brain pathology induced by Zika virus infection, independently of Oas1b, Journal of virology, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02445236

T. Z. Mann, L. B. Haddad, T. R. Williams, and S. L. Hills, Breast milk transmission of flaviviruses in the context of Zika virus: A systematic review, vol.32, pp.358-368, 2018.

N. J. Marchette, R. Garcia, and A. Rudnick, Isolation of Zika virus from Aedes aegypti mosquitoes in Malaysia. The American journal of tropical medicine and hygiene, vol.18, pp.411-415, 1969.

J. F. Marquis, R. Lacourse, L. Ryan, R. J. North, and P. Gros, Genetic and functional characterization of the mouse Trl3 locus in defense against tuberculosis, Journal of immunology, vol.182, pp.3757-3767, 1950.

A. L. Martin, M. D. Schwartz, S. C. Jameson, and Y. Shimizu, Selective regulation of CD8 effector T cell migration by the p110 gamma isoform of phosphatidylinositol 3-kinase, Journal of immunology, vol.180, pp.2081-2088, 1950.

R. B. Martines, J. Bhatnagar, A. M. De-oliveira-ramos, and H. P. Davi, Pathology of congenital Zika syndrome in Brazil: a case series, Lancet, vol.388, pp.898-904, 2016.

A. J. Martinot, P. Abbink, O. Afacan, and A. K. Prohl, Fetal Neuropathology in Zika Virus-Infected Pregnant Female Rhesus Monkeys, Cell, vol.173, p.1110, 2018.

T. Mashimo, M. Lucas, D. Simon-chazottes, and M. P. Frenkiel, A nonsense mutation in the gene encoding 2'-5'-oligoadenylate synthetase/L1 isoform is associated with West Nile virus susceptibility in laboratory mice, Proceedings of the National Academy of Sciences of the United States of America, vol.99, pp.11311-11316, 2002.

P. L. Maurizio, M. T. Ferris, G. R. Keele, and D. R. Miller, Bayesian Diallel Analysis Reveals Mx1-Dependent and Mx1-Independent Effects on Response to Influenza A Virus in Mice, Bethesda), vol.3, pp.427-445, 2018.

M. Mavigner, J. Raper, Z. Kovacs-balint, and S. Gumber, Postnatal Zika virus infection is associated with persistent abnormalities in brain structure, function, and behavior in infant macaques, Science translational medicine, p.10, 2018.

C. Mazeaud, W. Freppel, and L. Chatel-chaix, The Multiples Fates of the Flavivirus RNA Genome During Pathogenesis, Frontiers in genetics, vol.9, p.595, 2018.

M. K. Mccracken, G. D. Gromowski, H. L. Friberg, and X. Lin, Impact of prior flavivirus immunity on Zika virus infection in rhesus macaques, PLoS pathogens, vol.13, p.1006487, 2017.

F. Mcnab, K. Mayer-barber, A. Sher, A. Wack, and A. O&apos;garra, Type I interferons in infectious disease, Nat Rev Immunol, vol.15, pp.87-103, 2015.

P. S. Mead, N. K. Duggal, S. A. Hook, and M. Delorey, Zika Virus Shedding in Semen of Symptomatic Infected Men, N Engl J Med, vol.378, pp.1377-1385, 2018.

R. A. Medina and A. Garcia-sastre, Influenza A viruses: new research developments, Nat Rev Microbiol, vol.9, pp.590-603, 2011.

L. Meertens, A. Labeau, O. Dejarnac, and S. Cipriani, Axl Mediates ZIKA Virus Entry in Human Glial Cells and Modulates Innate Immune Responses, Cell reports, vol.18, pp.324-333, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02013511

M. S. Mehand, F. Al-shorbaji, P. Millett, and B. Murgue, The WHO R&D Blueprint: 2018 review of emerging infectious diseases requiring urgent research and development efforts, Antiviral Res, vol.159, pp.63-67, 2018.

K. C. Meier, C. L. Gardner, M. V. Khoretonenko, W. B. Klimstra, and K. D. Ryman, A mouse model for studying viscerotropic disease caused by yellow fever virus infection, PLoS pathogens, vol.5, p.1000614, 2009.

A. S. Melo, R. S. Aguiar, M. M. Amorim, and M. B. Arruda, Congenital Zika Virus Infection: Beyond Neonatal Microcephaly, JAMA neurology, vol.73, pp.1407-1416, 2016.

P. Mesci, A. Macia, S. M. Moore, and S. A. Shiryaev, Blocking Zika virus vertical transmission, Scientific reports, vol.8, p.1218, 2018.

I. Meyts, B. Bosch, A. Bolze, and B. Boisson, Exome and genome sequencing for inborn errors of immunity, The Journal of allergy and clinical immunology, vol.138, pp.957-969, 2016.

L. J. Miller, F. Nasar, C. W. Schellhase, and S. L. Norris, Zika Virus Infection in Syrian Golden Hamsters and Strain 13 Guinea Pigs. The American journal of tropical medicine and hygiene 98, pp.864-867, 2018.

T. C. Mills, A. Rautanen, K. S. Elliott, and T. Parks, IFITM3 and susceptibility to respiratory viral infections in the community, The Journal of infectious diseases, vol.209, pp.1028-1031, 2014.

J. J. Miner, B. Cao, J. Govero, and A. M. Smith, Zika Virus Infection during Pregnancy in Mice Causes Placental Damage and Fetal Demise, Cell, vol.165, pp.1081-1091, 2016.

J. J. Miner and M. S. Diamond, Zika Virus Pathogenesis and Tissue Tropism, vol.21, pp.134-142, 2017.

J. J. Miner, A. Sene, J. M. Richner, and A. M. Smith, Zika Virus Infection in Mice Causes Panuveitis with Shedding of Virus in Tears, Cell reports, vol.16, pp.3208-3218, 2016.

K. Modjarrad, L. Lin, S. L. George, and K. E. Stephenson, Preliminary aggregate safety and immunogenicity results from three trials of a purified inactivated Zika virus vaccine candidate: phase 1, randomised, double-blind, placebo-controlled clinical trials, Lancet, vol.391, pp.563-571, 2018.

E. L. Mohr, L. N. Block, C. M. Newman, and L. M. Stewart, Ocular and uteroplacental pathology in a macaque pregnancy with congenital Zika virus infection, PLoS One, vol.13, p.190617, 2018.

M. L. Moi, T. Nguyen, C. T. Nguyen, and T. Vu, Zika virus infection and microcephaly in Vietnam. The Lancet. Infectious diseases, vol.17, pp.805-806, 2017.

T. P. Monath and P. F. Vasconcelos, Journal of clinical virology : the official publication of the Pan American Society for, Clinical Virology, vol.64, pp.160-173, 2015.

C. A. Moore, J. E. Staples, W. B. Dobyns, and A. Pessoa, Characterizing the Pattern of Anomalies in Congenital Zika Syndrome for Pediatric Clinicians, JAMA pediatrics, vol.171, pp.288-295, 2017.

G. Morahan, L. Balmer, and D. Monley, Establishment of "The Gene Mine": a resource for rapid identification of complex trait genes, Mamm Genome, vol.19, pp.390-393, 2008.

E. M. Moresco and B. Beutler, Resisting viral infection: the gene by gene approach, Curr Opin Virol, vol.1, pp.513-518, 2011.

T. E. Morrison and M. S. Diamond, Animal Models of Zika Virus Infection, Pathogenesis, and Immunity, Journal of virology, p.91, 2017.

R. Mott and J. Flint, Dissecting quantitative traits in mice. Annual review of genomics and human genetics 14, pp.421-439, 2013.

I. J. Motta, B. R. Spencer, C. Da-silva, S. G. Arruda, and M. B. , Evidence for Transmission of Zika Virus by Platelet Transfusion, N Engl J Med, vol.375, pp.1101-1103, 2016.

P. Moussa, J. Marton, S. M. Vidal, and N. Fodil-cornu, Genetic dissection of NK cell responses, Front Immunol, vol.3, p.425, 2012.

S. Mukhopadhyay, R. J. Kuhn, and M. G. Rossmann, A structural perspective of the flavivirus life cycle, Nat Rev Microbiol, vol.3, pp.13-22, 2005.

U. Muller, U. Steinhoff, L. F. Reis, S. Hemmi, J. Pavlovic et al., Functional role of type I and type II interferons in antiviral defense, Science, vol.264, pp.1918-1921, 1994.

J. L. Munoz-jordan, Diagnosis of Zika Virus Infections: Challenges and Opportunities, The Journal of infectious diseases, vol.216, pp.951-956, 2017.

L. S. Munoz, B. Parra, and C. A. Pardo, Neurological Implications of Zika Virus Infection in Adults, The Journal of infectious diseases, vol.216, pp.897-905, 2017.

K. O. Murray, R. Gorchakov, A. R. Carlson, and R. Berry, Prolonged Detection of Zika Virus in Vaginal Secretions and Whole Blood, Emerging infectious diseases, vol.23, pp.99-101, 2017.

D. Musso and D. J. Gubler, Zika Virus, vol.29, pp.487-524, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02263641

D. Musso, T. Nhan, R. E. Roche, and C. , Potential for Zika virus transmission through blood transfusion demonstrated during an outbreak in French Polynesia, Euro surveillance : bulletin Europeen sur les maladies transmissibles = European communicable disease bulletin, p.19, 2013.

D. Musso, E. J. Nilles, and V. M. Cao-lormeau, Rapid spread of emerging Zika virus in the Pacific area. Clinical microbiology and infection : the official publication of the European Society of, Clinical Microbiology and Infectious Diseases, vol.20, pp.595-596, 2014.

D. Musso, C. Roche, R. E. Nhan, T. Teissier, A. Cao-lormeau et al., Potential sexual transmission of Zika virus, Emerging infectious diseases, vol.21, pp.359-361, 2015.

J. H. Nadeau and J. Auwerx, The virtuous cycle of human genetics and mouse models in drug discovery, Nature reviews. Drug discovery, vol.18, pp.255-272, 2019.

M. Nakahira, T. Tanaka, B. E. Robson, J. P. Mizgerd, and M. J. Grusby, Regulation of signal transducer and activator of transcription signaling by the tyrosine phosphatase PTP-BL, Immunity, vol.26, pp.163-176, 2007.

D. Nash, F. Mostashari, A. Fine, and J. Miller, The outbreak of West Nile virus infection in the New York City area in 1999, N Engl J Med, vol.344, pp.1807-1814, 2001.

T. Nedelko, H. Kollmus, F. Klawonn, and S. Spijker, Distinct gene loci control the host response to influenza H1N1 virus infection in a time-dependent manner, BMC genomics, vol.13, p.411, 2012.

C. J. Neufeldt, C. M. Acosta, E. G. Bartenschlager, and R. , Rewiring cellular networks by members of the Flaviviridae family, Nat Rev Microbiol, vol.16, pp.125-142, 2018.

M. J. Newport and C. Finan, Genome-wide association studies and susceptibility to infectious diseases, Briefings in functional genomics, vol.10, pp.98-107, 2011.

A. E. Ngono and S. Shresta, Immune Response to Dengue and Zika, Annual review of immunology, vol.36, pp.279-308, 2018.

H. N. Nguyen, X. Qian, H. Song, and G. L. Ming, Neural stem cells attacked by Zika virus, Cell research, vol.26, pp.753-754, 2016.

S. M. Nguyen, K. M. Antony, D. M. Dudley, and S. Kohn, Highly efficient maternal-fetal Zika virus transmission in pregnant rhesus macaques, PLoS pathogens, vol.13, p.1006378, 2017.

C. Nicolle, K. Saines, P. Brasil, T. Kerin, and Z. Vasconcelos, Delayed childhood neurodevelopment and neurosensory alterations in the second year of life in a prospective cohort of ZIKV-exposed children, Le destin des maladies infectieuses. Librairie Félix Alcan Nielsen, vol.25, pp.1213-1217, 1933.

K. E. Noll, M. T. Ferris, and M. T. Heise, The Collaborative Cross: A Systems Genetics Resource for Studying Host-Pathogen Interactions, Cell host & microbe, vol.25, pp.484-498, 2019.

E. Oehler, L. Watrin, P. Larre, and I. Leparc-goffart, Zika virus infection complicated by Guillain-Barre syndrome--case report, European communicable disease bulletin, p.19, 2013.

M. Ohno, Roles of eIF2alpha kinases in the pathogenesis of Alzheimer's disease, Frontiers in molecular neuroscience, vol.7, p.22, 2014.

A. S. Oliveira-melo, G. Malinger, R. Ximenes, P. O. Szejnfeld, A. Sampaio et al., Zika virus intrauterine infection causes fetal brain abnormality and microcephaly: tip of the iceberg? Ultrasound in obstetrics & gynecology : the official journal of the International Society, vol.47, pp.6-7, 2016.

D. Orofino, S. Passos, R. De-oliveira, and C. Farias, Cardiac findings in infants with in utero exposure to Zika virus-a cross sectional study, PLoS neglected tropical diseases, vol.12, p.6362, 2018.

C. E. Osuna, S. Y. Lim, C. Deleage, and B. D. Griffin, Zika viral dynamics and shedding in rhesus and cynomolgus macaques, Nature medicine, vol.22, pp.1448-1455, 2016.

E. S. Paixao, W. Y. Leong, L. C. Rodrigues, and A. Wilder-smith, Asymptomatic Prenatal Zika Virus Infection and Congenital Zika Syndrome, Open forum infectious diseases, vol.5, p.73, 2018.

C. Panayiotou, R. Lindqvist, C. Kurhade, and K. Vonderstein, Viperin Restricts Zika Virus and Tick-Borne Encephalitis Virus Replication by Targeting NS3 for Proteasomal Degradation, Journal of virology, p.92, 2018.

. Pano and . Paho, , 2018.

. Pano and . Paho, Epidemiological update. Reported increase of congenital microcephaly and other central nervous system symptoms, 2016.

P. Pantoja, E. X. Perez-guzman, I. V. Rodriguez, and L. J. White, Zika virus pathogenesis in rhesus macaques is unaffected by pre-existing immunity to dengue virus, Nature communications, vol.8, p.15674, 2017.

M. P. Papa, L. M. Meuren, S. Coelho, and C. Lucas, Zika Virus Infects, Activates, and Crosses Brain Microvascular Endothelial Cells, without Barrier Disruption, Front Microbiol, vol.8, p.2557, 2017.

R. D. Pardy, S. F. Valbon, and M. J. Richer, Running interference: Interplay between Zika virus and the host interferon response, Cytokine, vol.119, pp.7-15, 2019.

A. M. Paul, D. Acharya, L. Duty, and E. A. Thompson, Osteopontin facilitates West Nile virus neuroinvasion via neutrophil "Trojan horse, transport. Scientific reports, vol.7, p.4722, 2017.

A. M. Paul, D. Acharya, B. Neupane, and E. A. Thompson, Congenital Zika Virus Infection in Immunocompetent Mice Causes Postnatal Growth Impediment and Neurobehavioral Deficits, Front Microbiol, vol.9, p.2028, 2018.

A. A. Perelygin, S. V. Scherbik, I. B. Zhulin, B. M. Stockman, Y. Li et al., Positional cloning of the murine flavivirus resistance gene, Proceedings of the National Academy of Sciences of the United States of America, vol.99, pp.9322-9327, 2002.

A. Pessoa, V. Van-der-linden, M. Yeargin-allsopp, and M. Carvalho, Motor Abnormalities and Epilepsy in Infants and Children With Evidence of Congenital Zika Virus Infection, Pediatrics, vol.141, pp.167-179, 2018.

J. H. Pettersson, V. Eldholm, S. J. Seligman, and A. Lundkvist, How Did Zika Virus Emerge in the Pacific Islands and Latin America? mBio 7, 2016.

T. C. Pierson, Modeling antibody-enhanced dengue virus infection and disease in mice: protection or pathogenesis?, Cell host & microbe, vol.7, pp.85-86, 2010.

S. Plancoulaine, A. Gessain, P. Tortevoye, A. Boland-auge, A. Vasilescu et al., A major susceptibility locus for HTLV-1 infection in childhood maps to chromosome 6q27, Human molecular genetics, vol.15, pp.3306-3312, 2006.

L. Pomar, G. Malinger, G. Benoist, and G. Carles, Association between Zika virus and fetopathy: a prospective cohort study in French Guiana, Ultrasound in obstetrics & gynecology : the official journal of the International Society of Ultrasound in Obstetrics and Gynecology, vol.49, pp.729-736, 2017.

L. Pomar, M. Vouga, V. Lambert, and C. Pomar, Maternal-fetal transmission and adverse perinatal outcomes in pregnant women infected with Zika virus: prospective cohort study in French Guiana, BMJ, vol.363, p.4431, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01922382

P. Pound and M. Ritskes-hoitinga, Is it possible to overcome issues of external validity in preclinical animal research? Why most animal models are bound to fail, Journal of translational medicine, vol.16, p.304, 2018.

I. B. Rabe, J. E. Staples, J. Villanueva, and K. B. Hummel, Interim Guidance for Interpretation of Zika Virus Antibody Test Results, MMWR. Morbidity and mortality weekly report, vol.65, pp.543-546, 2016.

A. L. Rasmussen, A. Okumura, M. T. Ferris, and R. Green, Host genetic diversity enables Ebola hemorrhagic fever pathogenesis and resistance, Science, vol.346, pp.987-991, 2014.

W. Reed, Recent Researches concerning the Etiology, Propagation, and Prevention of Yellow Fever, by the United States Army Commission, The Journal of hygiene, vol.2, pp.101-119, 1902.

F. A. Rey, K. Stiasny, and F. X. Heinz, Flavivirus structural heterogeneity: implications for cell entry, Curr Opin Virol, vol.24, pp.132-139, 2017.

F. A. Rey, K. Stiasny, M. C. Vaney, M. Dellarole, and F. X. Heinz, The bright and the dark side of human antibody responses to flaviviruses: lessons for vaccine design, EMBO reports, vol.19, pp.206-224, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02346839

M. J. Ricciardi, D. M. Magnani, A. Grifoni, and Y. C. Kwon, Ontogeny of the B-and T-cell response in a primary Zika virus infection of a dengue-naive individual during the, PLoS neglected tropical diseases, vol.11, p.6000, 2016.

J. M. Richner and M. S. Diamond, Zika virus vaccines: immune response, current status, and future challenges, Curr Opin Immunol, vol.53, pp.130-136, 2018.

D. F. Robbiani, L. Bozzacco, J. R. Keeffe, and R. Khouri, Recurrent Potent Human Neutralizing Antibodies to Zika Virus in Brazil and Mexico, Cell, vol.169, pp.597-609, 2017.

A. Roberts, F. Pardo-manuel-de-villena, W. Wang, L. Mcmillan, and D. W. Threadgill, The polymorphism architecture of mouse genetic resources elucidated using genome-wide resequencing data: implications for QTL discovery and systems genetics, Mamm Genome, vol.18, pp.473-481, 2007.

A. R. Rogala, A. P. Morgan, A. M. Christensen, and T. J. Gooch, The Collaborative Cross as a resource for modeling human disease: CC011/Unc, a new mouse model for spontaneous colitis, Mamm Genome, vol.25, pp.95-108, 2014.

T. F. Rogers, E. C. Goodwin, B. Briney, and D. Sok, Zika virus activates de novo and crossreactive memory B cell responses in dengue-experienced donors, Science immunology, vol.2, 2017.

A. D. Rossi, F. R. Faucz, A. Melo, and P. Pezzuto, Variations in maternal adenylate cyclase genes are associated with congenital Zika syndrome in a cohort from Northeast, Brazil, Journal of internal medicine, vol.285, pp.215-222, 2019.

S. L. Rossi, G. D. Ebel, C. Shan, P. Y. Shi, and N. Vasilakis, Did Zika Virus Mutate to Cause Severe Outbreaks?, Trends in microbiology, vol.26, pp.877-885, 2018.

S. L. Rossi, R. B. Tesh, S. R. Azar, and A. E. Muruato, Characterization of a Novel Murine Model to Study Zika Virus, The American journal of tropical medicine and hygiene, vol.94, pp.1362-1369, 2016.

K. Ruchusatsawat, P. Wongjaroen, A. Posanacharoen, I. Rodriguez-barraquer, S. Sangkitporn et al., Long-term circulation of Zika virus in Thailand: an observational study, The Lancet. Infectious diseases, vol.19, pp.439-446, 2019.

C. Q. Sacramento, G. R. De-melo, C. S. De-freitas, and N. Rocha, The clinically approved antiviral drug sofosbuvir inhibits Zika virus replication, Scientific reports, vol.7, p.40920, 2017.

C. L. Sampieri and H. Montero, Breastfeeding in the time of Zika: a systematic literature review, PeerJ, vol.7, p.6452, 2019.

M. A. Samuel and M. S. Diamond, Alpha/beta interferon protects against lethal West Nile virus infection by restricting cellular tropism and enhancing neuronal survival, Journal of virology, vol.79, pp.13350-13361, 2005.

T. Sanada, G. Takaesu, R. Mashima, R. Yoshida, T. Kobayashi et al., FLN29 deficiency reveals its negative regulatory role in the Toll-like receptor (TLR) and retinoic acid-inducible gene I (RIG-I)-like helicase signaling pathway, The Journal of biological chemistry, vol.283, pp.33858-33864, 2008.

S. Cortes, M. Rivera, A. M. Yepez, M. Guimaraes, and C. V. , Clinical assessment and brain findings in a cohort of mothers, fetuses and infants infected with ZIKA virus, American journal of obstetrics and gynecology, vol.218, p.436, 2018.

G. Sapparapu, E. Fernandez, N. Kose, and C. Bin, Neutralizing human antibodies prevent Zika virus replication and fetal disease in mice, Nature, vol.540, pp.443-447, 2016.

W. Saron, A. Rathore, L. Ting, E. E. Ooi, J. Low et al., Flavivirus serocomplex cross-reactive immunity is protective by activating heterologous memory CD4 T cells, Science advances, vol.4, p.4297, 2018.

A. Satterfield-nash, K. Kotzky, J. Allen, and J. Bertolli, Health and Development at Age 19-24 Months of 19 Children Who Were Born with Microcephaly and Laboratory Evidence of Congenital Zika Virus Infection During the 2015 Zika Virus Outbreak -Brazil, MMWR. Morbidity and mortality weekly report, vol.66, pp.1347-1351, 2017.

M. C. Saul, V. M. Philip, L. G. Reinholdt, and E. J. Chesler, High-Diversity Mouse Populations for Complex Traits, Trends in Genetics, vol.35, pp.501-514, 2019.

G. Savidis, J. M. Perreira, J. M. Portmann, P. Meraner, Z. Guo et al., The IFITMs Inhibit Zika Virus Replication, Cell reports, vol.15, pp.2323-2330, 2016.

K. Schmitt, P. Charlins, M. Veselinovic, and L. Kinner-bibeau, Zika viral infection and neutralizing human antibody response in a BLT humanized mouse model, Virology, vol.515, pp.235-242, 2018.

W. M. Schneider, M. D. Chevillotte, and C. M. Rice, Interferon-Stimulated Genes: A Complex Web of Host Defenses, Annual review of immunology, vol.32, pp.513-545, 2014.

J. W. Schoggins, Interferon-stimulated genes: roles in viral pathogenesis, Curr Opin Virol, vol.6, pp.40-46, 2014.

K. Schughart and R. W. Williams, Systems Genetics, 2017.

J. M. Scott, T. J. Lebratti, J. M. Richner, and X. Jiang, Cellular and Humoral Immunity Protect against Vaginal Zika Virus Infection in Mice, Journal of virology, p.92, 2018.

M. Seferovic, C. Sanchez-san-martin, S. D. Tardif, and J. Rutherford, Experimental Zika Virus Infection in the Pregnant Common Marmoset Induces Spontaneous Fetal Loss and Neurodevelopmental Abnormalities, Scientific reports, vol.8, p.6851, 2018.

S. Shaily and A. Upadhya, Zika virus: Molecular responses and tissue tropism in the mammalian host, Reviews in medical virology, p.2050, 2019.

C. Shan, A. E. Muruato, B. W. Jagger, and J. Richner, A single-dose live-attenuated vaccine prevents Zika virus pregnancy transmission and testis damage, Nature communications, vol.8, p.676, 2017.

C. Shan, X. Xie, and P. Y. Shi, Zika Virus Vaccine: Progress and Challenges, Cell host & microbe, vol.24, pp.12-17, 2018.

Q. Shao, S. Herrlinger, S. L. Yang, F. Lai, J. M. Moore et al., Zika virus infection disrupts neurovascular development and results in postnatal microcephaly with brain damage, Development, vol.143, pp.4127-4136, 2016.

C. K. Shapiro-mendoza, M. E. Rice, R. R. Galang, and A. C. Fulton, Pregnancy Outcomes After Maternal Zika Virus Infection During Pregnancy, MMWR. Morbidity and mortality weekly report, vol.66, pp.615-621, 2016.

M. L. Sherer, P. Khanal, G. Talham, E. M. Brannick, M. S. Parcells et al., Zika virus infection of pregnant rats and associated neurological consequences in the offspring, PLoS One, vol.14, p.218539, 2019.

M. A. Sheridan, V. Balaraman, D. J. Schust, T. Ezashi, R. M. Roberts et al., African and Asian strains of Zika virus differ in their ability to infect and lyse primitive human placental trophoblast, PLoS One, vol.13, p.200086, 2018.

Y. Shi, S. Li, Q. Wu, and L. Sun, Vertical Transmission of the Zika Virus Causes Neurological Disorders in Mouse Offspring, Scientific reports, vol.8, p.3541, 2018.

S. A. Shiryaev, C. Farhy, A. Pinto, and C. T. Huang, Characterization of the Zika virus twocomponent NS2B-NS3 protease and structure-assisted identification of allosteric small-molecule antagonists, Antiviral Res, vol.143, pp.218-229, 2017.

S. A. Shiryaev, P. Mesci, A. Pinto, and I. Fernandes, Repurposing of the anti-malaria drug chloroquine for Zika Virus treatment and prophylaxis, Scientific reports, vol.7, p.15771, 2017.

J. R. Shorter, F. Odet, D. L. Aylor, and W. Pan, Male Infertility Is Responsible for Nearly Half of the Extinction Observed in the Mouse Collaborative Cross, Genetics, vol.206, pp.557-572, 2017.

S. Shresta, J. L. Kyle, H. M. Snider, M. Basavapatna, P. R. Beatty et al., Interferon-dependent immunity is essential for resistance to primary dengue virus infection in mice, whereas T-and B-celldependent immunity are less critical, Journal of virology, vol.78, pp.2701-2710, 2004.

S. Shresta, K. L. Sharar, D. M. Prigozhin, P. R. Beatty, and E. Harris, Murine model for dengue virusinduced lethal disease with increased vascular permeability, Journal of virology, vol.80, pp.10208-10217, 2006.

Y. Simonin, N. Erkilic, K. Damodar, and M. Cle, Zika virus induces strong inflammatory responses and impairs homeostasis and function of the human retinal pigment epithelium, EBioMedicine, vol.39, pp.315-331, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02083544

R. K. Singh, K. Dhama, R. Khandia, and A. Munjal, Prevention and Control Strategies to Counter Zika Virus, a Special Focus on Intervention Approaches against Vector Mosquitoes-Current Updates, Front Microbiol, vol.9, p.87, 2018.

D. Sirohi and R. J. Kuhn, Zika Virus Structure, Maturation, and Receptors, vol.216, pp.935-944, 2017.

T. L. Smallwood, D. M. Gatti, P. Quizon, and G. M. Weinstock, High-resolution genetic mapping in the diversity outbred mouse population identifies Apobec1 as a candidate gene for atherosclerosis, Bethesda), vol.3, pp.2353-2363, 2014.

D. R. Smith, B. Hollidge, S. Daye, and X. Zeng, Neuropathogenesis of Zika Virus in a Highly Susceptible Immunocompetent Mouse Model after Antibody Blockade of Type I Interferon, PLoS neglected tropical diseases, vol.11, p.5296, 2017.

D. R. Smith, T. R. Sprague, B. S. Hollidge, and S. M. Valdez, African and Asian Zika Virus Isolates Display Phenotypic Differences Both In Vitro and In Vivo, The American journal of tropical medicine and hygiene, vol.98, pp.432-444, 2018.

A. Snyder-keller, L. Kramer, S. Zink, and V. J. Bolivar, Mouse strain and sex-dependent differences in long-term behavioral abnormalities and neuropathologies after developmental zika infection, The Journal of neuroscience : the official journal of the Society for Neuroscience, vol.39, pp.5393-5403, 2019.

A. Soriano-arandes, I. Rivero-calle, E. Nastouli, M. Espiau, M. A. Frick et al., What we know and what we don't know about perinatal Zika virus infection: a systematic review, Expert review of anti-infective therapy, vol.16, pp.243-254, 2018.

K. R. Spindler, L. Fang, M. L. Moore, G. N. Hirsch, C. C. Brown et al., SJL/J mice are highly susceptible to infection by mouse adenovirus type 1, Journal of virology, vol.75, pp.12039-12046, 2001.

H. Spits, J. H. Bernink, and L. Lanier, NK cells and type 1 innate lymphoid cells: partners in host defense, Nature immunology, vol.17, pp.758-764, 2016.

A. Srivastava, A. P. Morgan, M. L. Najarian, and V. K. Sarsani, Genomes of the Mouse Collaborative Cross, Genetics, vol.206, pp.537-556, 2017.

A. C. Stabell, N. R. Meyerson, R. C. Gullberg, and A. R. Gilchrist, Dengue viruses cleave STING in humans but not in nonhuman primates, their presumed natural reservoir, 2018.

P. Staeheli, R. Grob, E. Meier, J. G. Sutcliffe, and O. Haller, Influenza virus-susceptible mice carry Mx genes with a large deletion or a nonsense mutation, Molecular and cellular biology, vol.8, pp.4518-4523, 1988.

P. Staeheli, O. Haller, W. Boll, J. Lindenmann, and C. Weissmann, Mx protein: constitutive expression in 3T3 cells transformed with cloned Mx cDNA confers selective resistance to influenza virus, Cell, vol.44, pp.147-158, 1986.

P. Staeheli, D. Pravtcheva, L. G. Lundin, M. Acklin, F. Ruddle et al., Interferon-regulated influenza virus resistance gene Mx is localized on mouse chromosome 16, Journal of virology, vol.58, pp.967-969, 1986.

K. Stettler, M. Beltramello, D. A. Espinosa, and V. Graham, Specificity, cross-reactivity, and function of antibodies elicited by Zika virus infection, Science, vol.353, pp.823-826, 2016.

M. Suffiotti, S. J. Carmona, C. Jandus, and D. Gfeller, Identification of innate lymphoid cells in singlecell RNA-Seq data, Immunogenetics, vol.69, pp.439-450, 2017.

K. L. Svenson, D. M. Gatti, W. Valdar, and C. E. Welsh, High-resolution genetic mapping using the Mouse Diversity outbred population, Genetics, vol.190, pp.437-447, 2012.

S. Swaminathan, R. Schlaberg, J. Lewis, K. E. Hanson, and M. R. Couturier, Fatal Zika Virus Infection with Secondary Nonsexual Transmission, N Engl J Med, vol.375, pp.1907-1909, 2016.

J. A. Swanstrom, J. A. Plante, K. S. Plante, and E. F. Young, Dengue Virus Envelope Dimer Epitope Monoclonal Antibodies Isolated from Dengue Patients Are Protective against Zika Virus, 2016.

F. M. Szaba, M. Tighe, L. W. Kummer, and K. G. Lanzer, Zika virus infection in immunocompetent pregnant mice causes fetal damage and placental pathology in the absence of fetal infection, PLoS pathogens, vol.14, p.1006994, 2018.

C. Talero-gutierrez, A. Rivera-molina, C. Perez-pavajeau, I. Ossa-ospina, C. Santos-garcia et al., Zika virus epidemiology: from Uganda to world pandemic, an update, Epidemiology and infection, pp.1-7, 2018.

H. Tang, C. Hammack, S. C. Ogden, and Z. Wen, Zika Virus Infects Human Cortical Neural Progenitors and Attenuates Their Growth, Cell stem cell, vol.18, pp.587-590, 2016.

W. W. Tang, M. P. Young, A. Mamidi, J. A. Regla-nava, K. Kim et al., A Mouse Model of Zika Virus Sexual Transmission and Vaginal Viral Replication, Cell reports, vol.17, pp.3091-3098, 2016.

B. A. Taylor, Recombinant Inbred Strains: Use in Gene Mapping, pp.423-438, 1978.

P. Tebas, C. C. Roberts, K. Muthumani, and E. L. Reuschel, Safety and Immunogenicity of an Anti-Zika Virus DNA Vaccine -Preliminary Report, N Engl J Med The Jackson Laboratory, 2017.

M. Theiler, SUSCEPTIBILITY OF WHITE MICE TO THE VIRUS OF YELLOW FEVER, Science, vol.71, p.367, 1930.

D. W. Threadgill, K. W. Hunter, and R. W. Williams, Genetic dissection of complex and quantitative traits: from fantasy to reality via a community effort, Mamm Genome, vol.13, pp.175-178, 2002.

S. Tripathi, V. R. Balasubramaniam, J. A. Brown, and I. Mena, A novel Zika virus mouse model reveals strain specific differences in virus pathogenesis and host inflammatory immune responses, PLoS pathogens, vol.13, p.1006258, 2017.

L. Turtle and T. Solomon, Japanese encephalitis -the prospects for new treatments, Nat Rev Neurol, vol.14, pp.298-313, 2018.

, UNC Systems Genetics -The Collaborative Cross, UNC Systems Genetics

R. Uraki, J. Hwang, K. A. Jurado, and S. Householder, Zika virus causes testicular atrophy, Science advances, vol.3, p.1602899, 2017.

N. Urosevic, J. P. Mansfield, J. S. Mackenzie, and G. R. Shellam, Low resolution mapping around the flavivirus resistance locus (Flv) on mouse chromosome 5, Mamm Genome, vol.6, pp.454-458, 1995.

J. F. Valdes-lopez, P. A. Velilla, and S. Urcuqui-inchima, Chikungunya Virus and Zika Virus, Two Different Viruses Examined with a Common Aim: Role of Pattern Recognition Receptors on the Inflammatory Response, Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research, vol.39, pp.507-521, 2019.

G. C. Valentine, M. D. Seferovic, S. W. Fowler, and A. M. Major, Timing of Gestational Exposure to Zika Virus is Associated with Postnatal Growth Restriction in a Murine Model, American journal of obstetrics and gynecology, vol.219, p.409, 2018.

M. F. Van-den-broek, U. Muller, S. Huang, R. M. Zinkernagel, and M. Aguet, Immune defence in mice lacking type I and/or type II interferon receptors, Immunological reviews, vol.148, pp.5-18, 1995.

K. H. Van-der-hoek, N. S. Eyre, B. Shue, and O. Khantisitthiporn, Viperin is an important host restriction factor in control of Zika virus infection, Scientific reports, vol.7, p.4475, 2017.

H. Van-der-linden, J. Carvalho, M. D. Van-der-linden, V. Lacerda, and K. M. , Epilepsy Profile in Infants with Congenital Zika Virus Infection, N Engl J Med, vol.379, pp.891-892, 2018.

V. Van-der-linden, A. Pessoa, W. Dobyns, and A. J. Barkovich, With Congenital Zika Virus Infection Without Microcephaly at Birth -Brazil, MMWR. Morbidity and mortality weekly report, vol.65, pp.1343-1348, 2015.

H. B. Van-der-worp, D. W. Howells, E. S. Sena, M. J. Porritt, S. Rewell et al., Can animal models of disease reliably inform human studies?, PLoS medicine, vol.7, p.1000245, 2010.

F. Varenne, Epistémologie des modèles et des simulations, Epistémologie des modèles et des simulations : tour d'horizon et tendances, 2008.

A. Venkatratnam, J. S. House, K. Konganti, and C. Mckenney, Population-based dose-response analysis of liver transcriptional response to trichloroethylene in mouse, Mamm Genome, vol.29, pp.168-181, 2018.

K. Vered, C. Durrant, R. Mott, and F. A. Iraqi, Susceptibility to Klebsiella pneumonaie infection in collaborative cross mice is a complex trait controlled by at least three loci acting at different time points, BMC genomics, vol.15, p.865, 2014.

M. S. Vermillion, J. Lei, Y. Shabi, and V. K. Baxter, Intrauterine Zika virus infection of pregnant immunocompetent mice models transplacental transmission and adverse perinatal outcomes, Nature communications, vol.8, p.14575, 2017.

R. Vianna, K. L. Lovero, S. A. Oliveira, and A. R. Fernandes, Children Born to Mothers with Rash During Zika Virus Epidemic in Brazil: First 18 Months of Life, Journal of tropical pediatrics, 2019.

M. A. Vieira, A. P. Romano, A. S. Borba, and E. V. Silva, West Nile Virus Encephalitis: The First Human Case Recorded in Brazil, The American journal of tropical medicine and hygiene, vol.93, pp.377-379, 2015.

K. Vonderstein, E. Nilsson, P. Hubel, N. Skalman, and L. , Viperin Targets Flavivirus Virulence by Inducing Assembly of Noninfectious Capsid Particles, Journal of virology, p.92, 2018.

C. L. Walker, M. E. Little, J. A. Roby, and B. Armistead, Zika virus and the nonmicrocephalic fetus: why we should still worry, American journal of obstetrics and gynecology, vol.220, pp.45-56, 2019.

C. L. Walker, A. A. Merriam, E. O. Ohuma, and M. K. Dighe, Femur-sparing pattern of abnormal fetal growth in pregnant women from New York City after maternal Zika virus infection, American journal of obstetrics and gynecology, vol.219, pp.181-187, 2018.

L. F. Wang and D. E. Anderson, Viruses in bats and potential spillover to animals and humans, Curr Opin Virol, vol.34, pp.79-89, 2019.

S. Wang, S. Hong, Y. Q. Deng, and Q. Ye, Transfer of convalescent serum to pregnant mice prevents Zika virus infection and microcephaly in offspring, Cell research, vol.27, pp.158-160, 2017.

S. C. Weaver, Urbanization and geographic expansion of zoonotic arboviral diseases: mechanisms and potential strategies for prevention, Trends in microbiology, vol.21, pp.360-363, 2013.

J. Wen, W. W. Tang, N. Sheets, J. Ellison, A. Sette et al., Identification of Zika virus epitopes reveals immunodominant and protective roles for dengue virus cross-reactive CD8+ T cells, Nature microbiology, vol.2, p.17036, 2017.

W. Schreur, P. J. Van-keulen, L. Anjema, D. Kant, J. Kortekaas et al., Microencephaly in fetal piglets following in utero inoculation of Zika virus, Emerging microbes & infections, vol.7, p.42, 2018.

A. Wilder-smith, E. E. Ooi, O. Horstick, and B. Wills, Dengue. Lancet, vol.393, pp.350-363, 2019.

E. G. Williams and J. Auwerx, The Convergence of Systems and Reductionist Approaches in Complex Trait Analysis, Cell, vol.162, pp.23-32, 2015.

C. W. Winkler, L. M. Myers, T. A. Woods, and R. J. Messer, Adaptive Immune Responses to Zika Virus Are Important for Controlling Virus Infection and Preventing Infection in Brain and Testes, Journal of immunology, vol.198, pp.3526-3535, 1950.

C. W. Winkler and K. E. Peterson, Using immunocompromised mice to identify mechanisms of Zika virus transmission and pathogenesis, Immunology, 2017.

C. W. Winkler, T. A. Woods, R. Rosenke, D. P. Scott, S. M. Best et al., Sexual and Vertical Transmission of Zika Virus in anti-interferon receptor-treated Rag1-deficient mice, Scientific reports, vol.7, p.7176, 2017.

F. C. Wong and Y. M. Lo, Prenatal Diagnosis Innovation: Genome Sequencing of Maternal Plasma, Annu Rev Med, vol.67, pp.419-432, 2016.

, World Health Organization, 2016.

, World Health Organization (15 February 2018) Zika virus (ZIKV) classification table

, World Health Organization (2012) Handbook for integrated vector management

, World Health Organization, Infant feeding in areas of Zika virus transmission

, World Health Organization, WHO vaccine trial tracker

, United Nations International Children's Emergency Fund, Zika Virus (ZIKV) Vaccine Target Product Profile (TPP): Vaccine to protect against congenital Zika syndrome for use during an emergency

K. Y. Wu, G. L. Zuo, X. F. Li, and Q. Ye, Vertical transmission of Zika virus targeting the radial glial cells affects cortex development of offspring mice, Cell research, vol.26, pp.645-654, 2016.

Y. Wu, Q. Liu, J. Zhou, and W. Xie, Zika virus evades interferon-mediated antiviral response through the co-operation of multiple nonstructural proteins in vitro, Cell discovery, vol.3, p.17006, 2017.

H. Xia, H. Luo, C. Shan, and A. E. Muruato, An evolutionary NS1 mutation enhances Zika virus evasion of host interferon induction, Nature communications, vol.9, p.414, 2018.

D. Xu, C. Li, C. F. Qin, and Z. Xu, Update on the Animal Models and Underlying Mechanisms for ZIKV-Induced Microcephaly, Annual review of virology, vol.6, pp.459-479, 2019.

P. D. Yadav, B. Malhotra, G. Sapkal, and D. A. Nyayanit, Zika virus outbreak in Rajasthan, India in 2018 was caused by a virus endemic to Asia. Infection, genetics and evolution : journal of molecular epidemiology and evolutionary genetics in infectious diseases, vol.69, pp.199-202, 2019.

D. Yang, N. L. Li, D. Wei, and B. Liu, The E3 ligase TRIM56 is a host restriction factor of Zika virus and depends on its RNA-binding activity but not miRNA regulation, for antiviral function, PLoS neglected tropical diseases, vol.13, p.7537, 2019.

L. J. Yockey, L. Varela, T. Rakib, and W. Khoury-hanold, Vaginal Exposure to Zika Virus during Pregnancy Leads to Fetal Brain Infection, Cell, vol.166, pp.1247-1256, 1244.

L. Yuan, X. Y. Huang, Z. Y. Liu, and F. Zhang, A single mutation in the prM protein of Zika virus contributes to fetal microcephaly, Science, vol.358, pp.933-936, 2017.

C. Zanluca, V. C. Melo, A. L. Mosimann, G. I. Santos, C. N. Santos et al., First report of autochthonous transmission of Zika virus in Brazil, Memorias do Instituto Oswaldo Cruz, vol.110, pp.569-572, 2015.

C. J. Zeiss, D. M. Gatti, O. Toro-salazar, and C. Davis, Doxorubicin-Induced Cardiotoxicity in Collaborative Cross (CC) Mice Recapitulates Individual Cardiotoxicity in Humans, Bethesda), vol.3, pp.2637-2646, 2019.

B. Zhang, Y. Y. Lin, M. Dai, and Y. Zhuang, Id3 and Id2 act as a dual safety mechanism in regulating the development and population size of innate-like gammadelta T cells, Journal of immunology, vol.192, pp.1055-1063, 1950.

J. Zhang, D. Malo, R. Mott, J. J. Panthier, X. Montagutelli et al., Identification of new loci involved in the host susceptibility to Salmonella Typhimurium in collaborative cross mice, BMC genomics, vol.19, p.303, 2018.

J. Zhang, M. Teh, J. Kim, and M. M. Eva, A loss-of-function mutation in Itgal contributes to the high susceptibility of Collaborative Cross strain CC042 to Salmonella infections, 2019.

S. Y. Zhang, E. Jouanguy, S. Ugolini, and A. Smahi, TLR3 deficiency in patients with herpes simplex encephalitis, Science, vol.317, pp.1522-1527, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00297308

Y. H. Zhang, Y. Zhao, N. Li, and Y. C. Peng, Interferon-induced transmembrane protein-3 genetic variant rs12252-C is associated with severe influenza in Chinese individuals, Nature communications, vol.4, p.1418, 2013.

H. Zhao, E. Fernandez, K. A. Dowd, and S. D. Speer, Structural Basis of Zika Virus-Specific Antibody Protection, Cell, vol.166, pp.1016-1027, 2016.

X. Zhu, C. Li, S. K. Afridi, and S. Zu, E90 subunit vaccine protects mice from Zika virus infection and microcephaly, Acta neuropathologica communications, vol.6, p.77, 2018.

S. Zompi, B. H. Santich, P. R. Beatty, and E. Harris, Protection from secondary dengue virus infection in a mouse model reveals the role of serotype cross-reactive B and T cells, Journal of immunology, vol.188, pp.404-416, 1950.