L. B. Ivashkiv and L. T. Donlin, Regulation of type I interferon responses, Nat. Rev. Immunol, vol.14, pp.36-49, 2014.

X. Dagenais-lussier, H. Loucif, A. Murira, X. Laulhé, S. Stäger et al., Seustained IFN-I expression during established persistent viral infection: A "bad seed" for protective immunity, Viruses, vol.10, p.12, 2017.

M. P. Rodero and Y. J. Crow, Type I interferon-mediated monogenic autoinflammation: The type I interferonopathies, a conceptual overview, J. Exp. Med, vol.213, pp.2527-2538, 2016.

R. Banchereau, S. Hong, B. Cantarel, N. Baldwin, J. Baisch et al., Personalized immunomonitoring uncovers molecular networks that stratify lupus patients, Cell, vol.165, pp.551-565, 2016.

M. P. Rodero, J. Decalf, V. Bondet, D. Hunt, G. I. Rice et al., Detection of interferon alpha protein reveals differential levels and cellular sources in disease, J. Exp. Med, vol.214, pp.1547-1555, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-01534181

G. Grouard, M. Rissoan, L. Filgueira, I. Durand, J. Banchereau et al., The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand

, J. Exp. Med, vol.185, pp.1101-1111, 1997.

M. Colonna and M. Cella, Crosspresentation: Plasmacytoid dendritic cells are in the business, Immunity, vol.27, pp.419-421, 2007.

F. P. Siegal, N. Kadowaki, M. Shodell, P. A. Fitzgerald-bocarsly, K. Shah et al., The nature of the principal type I interferon-producing cells in human blood, Science, vol.284, pp.1835-1837, 1999.

D. Jarrossay, G. Napolitani, M. Colonna, F. Sallusto, and A. Lanzavecchia, Specialization and complementarity in microbial molecule recognition by human myeloid and plasmacytoid dendritic cells, Eur. J. Immunol, vol.31, pp.3388-3393, 2001.

N. Kadowaki and Y. Liu, Natural type I interferon-producing cells as a link between innate and adaptive immunity, Hum. Immunol, vol.63, pp.1126-1132, 2002.

M. Gandini, C. Gras, E. L. Azeredo, L. M. De-oliveira-pinto, N. Smith et al., Dengue virus activates membrane TRAIL relocalization and IFN-? production by human plasmacytoid dendritic cells in vitro and in vivo, PLOS Negl. Trop. Dis, vol.7, p.2257, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00844933

A. W. Hardy, D. R. Graham, G. M. Shearer, and J. P. , HIV turns plasmacytoid dendritic cells (pDC) into TRAIL-expressing killer pDC and down-regulates HIV coreceptors by Toll-like receptor 7-induced IFN-alpha, Proc. Natl. Acad. Sci. U.S.A, vol.104, pp.17453-17458, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00177945

H. Hemmi, O. Takeuchi, T. Kawai, T. Kaisho, S. Sato et al., A Toll-like receptor recognizes bacterial DNA, Nature, vol.408, pp.740-745, 2000.

M. Swiecki and M. Colonna, The multifaceted biology of plasmacytoid dendritic cells, Nat. Rev. Immunol, vol.15, pp.471-485, 2015.

L. Rönnblom and V. , The innate immune system in SLE: Type I interferons and dendritic cells, Lupus, vol.17, pp.394-399, 2008.

K. Sakata, S. Nakayamada, Y. Miyazaki, S. Kubo, A. Ishii et al., Up-regulation of TLR7-mediated IFN-? production by plasmacytoid dendritic cells in patients with systemic lupus erythematosus, Front. Immunol, vol.9, p.1957, 2018.

B. F. Chong and C. Mohan, Targeting the CXCR4/CXCL12 axis in systemic lupus erythematosus, Expert Opin. Ther. Targets, vol.13, pp.1147-1153, 2009.

A. Wang, A. Fairhurst, K. Tus, S. Subramanian, Y. Liu et al., CXCR4/CXCL12 hyperexpression plays a pivotal role in the pathogenesis of lupus, J. Immunol, vol.182, pp.4448-4458, 2009.

N. Smith, N. Pietrancosta, S. Davidson, J. Dutrieux, L. Chauveau et al., Natural amines inhibit activation of human plasmacytoid dendritic cells through CXCR4 engagement, Nat. Commun, vol.8, p.14253, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02075366

B. Wu, E. Y. Chien, C. D. Mol, G. Fenalti, W. Liu et al., Structures of the CXCR4 chemokine GPCR with small-molecule and cyclic peptide antagonists, Science, vol.330, pp.1066-1071, 2010.

J. F. Dipersio, I. N. Micallef, P. J. Stiff, B. J. Bolwell, R. T. Maziarz et al., Phase III prospective randomized double-blind placebo-controlled trial of plerixafor plus granulocyte colony-stimulating factor compared with placebo plus granulocyte colony-stimulating factor for autologous stem-cell mobilization and transplantation for patients with non-hodgkin's lymphoma, J. Clin. Oncol, vol.27, pp.4767-4773, 2009.

D. J. Scholten, M. Canals, D. Maussang, L. Roumen, M. J. Smit et al., Pharmacological modulation of chemokine receptor function, Br. J. Pharmacol, vol.165, pp.1617-1643, 2012.

M. M. Rosenkilde, L. Gerlach, S. Hatse, R. T. Skerlj, D. Schols et al., Molecular mechanism of action of monocyclam versus bicyclam non-peptide antagonists in the CXCR4 chemokine receptor, J. Biol. Chem, vol.282, pp.27354-27365, 2007.

M. M. Rosenkilde, L. Gerlach, J. S. Jakobsen, R. T. Skerlj, G. J. Bridger et al., Molecular mechanism of AMD3100 antagonism in the CXCR4 receptor: Transfer of binding site to the CXCR3 receptor, J. Biol. Chem, vol.279, pp.3033-3041, 2004.

G. A. Donzella, D. Schols, S. W. Lin, J. A. Esté, K. A. Nagashima et al., AMD3100, a small molecule inhibitor of HIV-1 entry via the CXCR4 co-receptor, Nat. Med, vol.4, pp.72-77, 1998.

G. Thoma, M. B. Streiff, J. Kovarik, F. Glickman, T. Wagner et al., Orally bioavailable isothioureas block function of the chemokine receptor CXCR4 in vitro and in vivo, J. Med. Chem, vol.51, pp.7915-7920, 2008.

N. Smith, P. O. Vidalain, S. Nisole, and J. P. , An efficient method for gene silencing in human primary plasmacytoid dendritic cells: Silencing of the TLR7/IRF-7 pathway as a proof of concept, Sci. Rep, vol.6, p.29891, 2016.

J. P. Herbeuval, J. Nilsson, A. Boasso, A. W. Hardy, M. J. Kruhlak et al., Differential expression of IFN-alpha and TRAIL/DR5 in lymphoid tissue of progressor versus nonprogressor HIV-1-infected patients, Proc. Natl. Acad. Sci. U.S.A, vol.103, pp.7000-7005, 2006.

J. Herbeuval, A. Boasso, J. Grivel, A. W. Hardy, S. A. Anderson et al., TNF-related apoptosis-inducing ligand (TRAIL) in HIV-1-infected patients and its in vitro production by antigen-presenting cells, Blood, vol.105, pp.2458-2464, 2005.

A. Kaul, C. Gordon, M. K. Crow, Z. Touma, M. B. Urowitz et al., Systemic lupus erythematosus, Nat. Rev. Dis. Primers, vol.2, p.16039, 2016.

V. Salvi, V. Gianello, S. Busatto, P. Bergese, L. Andreoli et al., Exosome-delivered microRNAs promote IFN-alpha secretion by human plasmacytoid DCs via TLR7, JCI Insight, vol.3, p.98204, 2018.

G. Murayama, N. Furusawa, A. Chiba, K. Yamaji, N. Tamura et al., Enhanced IFN-alpha production is associated with increased TLR7 retention in the lysosomes of palasmacytoid dendritic cells in systemic lupus erythematosus, Arthritis Res. Ther, vol.19, p.234, 2017.

V. Sisirak, D. Ganguly, K. L. Lewis, C. Couillault, L. Tanaka et al., Genetic evidence for the role of plasmacytoid dendritic cells in systemic lupus erythematosus, J. Exp. Med, vol.211, pp.1969-1976, 2014.

C. Guiducci, M. Gong, Z. Xu, M. Gill, D. Chaussabel et al., TLR recognition of self nucleic acids hampers glucocorticoid activity in lupus, Nature, vol.465, pp.937-941, 2010.

H. Zhuang, C. Szeto, S. Han, L. Yang, and W. H. Reeves, Animal models of interferon signature positive lupus, Front. Immunol, vol.6, p.291, 2015.

E. Savarese, C. Steinberg, R. D. Pawar, W. Reindl, S. Akira et al., Requirement of Toll-like receptor 7 for pristane-induced production of autoantibodies and development of murine lupus nephritis, Arthritis Rheum, vol.58, pp.1107-1115, 2008.

X. Chen, R. Cui, R. Li, H. Lin, Z. Huang et al., Development of pristane induced mice model for lupus with atherosclerosis and analysis of TLR expression, Clin. Exp. Rheumatol, vol.34, pp.600-608, 2016.

M. Lucas-hourani, D. Dauzonne, P. Jorda, G. Cousin, A. Lupan et al., Inhibition of pyrimidine biosynthesis pathway suppresses viral growth through innate immunity, PLOS Pathog, vol.9, p.1003678, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-01113535

M. J. Mirzayan, R. E. Schmidt, and T. Witte, Prognostic parameters for flare in systemic lupus erythematosus, Rheumatology, vol.39, pp.1316-1319, 2000.

S. Caielli, S. Athale, B. Domic, E. Murat, M. Chandra et al., Oxidized mitochondrial nucleoids released by neutrophils drive type I interferon production in human lupus, J. Exp. Med, vol.213, pp.697-713, 2016.

S. S. Diebold, T. Kaisho, H. Hemmi, S. Akira, and C. Reis-e-sousa, Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA, Science, vol.303, pp.1529-1531, 2004.

S. S. Diebold, M. Montoya, H. Unger, L. Alexopoulou, P. Roy et al., Viral infection switches non-plasmacytoid dendritic cells into high interferon producers, Nature, vol.424, pp.324-328, 2003.

J. Banchereau and V. , Type I interferon in systemic lupus erythematosus and other autoimmune diseases, Immunity, vol.25, pp.383-392, 2006.

T. B. Niewold, Type I interferon in human autoimmunity. Front. Immunol, vol.5, p.306, 2014.

L. Cheng, J. Ma, J. Li, D. Li, G. Li et al., Blocking type I interferon signaling enhances T cell recovery and reduces HIV-1 reservoirs, J. Clin. Invest, vol.127, pp.269-279, 2017.

S. G. Deeks, P. M. Odorizzi, and R. P. Sekaly, The interferon paradox: Can inhibiting an antiviral mechanism advance an HIV cure?, J. Clin. Invest, vol.127, pp.103-105, 2017.

A. Zhen, V. Rezek, C. Youn, B. Lam, N. Chang et al., Targeting type I interferon-mediated activation restores immune function in chronic HIV infection, J. Clin. Invest, vol.127, pp.260-268, 2017.

S. Luo, Y. Wang, M. Zhao, and Q. Lu, The important roles of type I interferon and interferon-inducible genes in systemic lupus erythematosus, Int. Immunopharmacol, vol.40, pp.542-549, 2016.

L. Bennett, A. K. Palucka, E. Arce, V. Cantrell, J. Borvak et al., Interferon and granulopoiesis signatures in systemic lupus erythematosus blood, J. Exp. Med, vol.197, pp.711-723, 2003.

V. Pascual, J. Banchereau, and A. K. Palucka, The central role of dendritic cells and interferon-alpha in SLE, Curr. Opin. Rheumatol, vol.15, pp.548-556, 2003.

R. Felten, E. Dervovic, F. Chasset, J. E. Gottenberg, J. Sibilia et al., The 2018 pipeline of targeted therapies under clinical development for systemic lupus erythematosus: A systematic review of trials, Autoimmun. Rev, vol.17, pp.781-790, 2018.

S. L. Rowland, J. M. Riggs, S. Gilfillan, M. Bugatti, W. Vermi et al., Early, transient depletion of plasmacytoid dendritic cells ameliorates autoimmunity in a lupus model, J. Exp. Med, vol.211, pp.1977-1991, 2014.

M. Hasan, N. Dobbs, S. Khan, M. A. White, E. K. Wakeland et al., Cutting edge: Inhibiting TBK1 by compound II ameliorates autoimmune disease in mice, J. Immunol, vol.195, pp.4573-4577, 2015.

H. Zhuang, S. Han, Y. Xu, Y. Li, H. Wang et al., Toll-like receptor 7-stimulated tumor necrosis factor ? causes bone marrow damage in systemic lupus erythematosus, Arthritis Rheum, vol.66, pp.140-151, 2014.

P. Y. Lee, J. S. Weinstein, D. C. Nacionales, P. O. Scumpia, Y. Li et al., A novel type I IFN-producing cell subset in murine lupus, J. Immunol, vol.180, pp.5101-5108, 2008.

B. Debnath, S. Xu, F. Grande, A. Garofalo, and N. Neamati, Small molecule inhibitors of CXCR4. Theranostics, vol.3, pp.47-75, 2013.

R. K. Mishra, A. K. Shum, L. C. Platanias, R. J. Miller, and G. E. Schiltz, Discovery and characterization of novel small-molecule CXCR4 receptor agonists and antagonists, Sci. Rep, vol.6, p.30155, 2016.

K. Nuji?, M. Banjanac, V. Muni?, D. Polan?ec, and V. E. Haber, Impairment of lysosomal functions by azithromycin and chloroquine contributes to anti-inflammatory phenotype, Cell. Immunol, vol.279, pp.78-86, 2012.

S. Meyer, M. Woodward, C. Hertel, P. Vlaicu, Y. Haque et al., AIRE-deficient patients harbor unique high-affinity disease-ameliorating autoantibodies, Cell, vol.166, pp.582-595, 2016.

T. Sterling and J. J. Irwin, ZINC 15-Ligand discovery for everyone, J. Chem. Inf. Model, vol.55, pp.2324-2337, 2015.

H. Zhao and A. Caflisch, Discovery of ZAP70 inhibitors by high-throughput docking into a conformation of its kinase domain generated by molecular dynamics, Bioorg. Med. Chem. Lett, vol.23, pp.5721-5726, 2013.

J. C. Phillips, R. Braun, W. Wang, J. Gumbart, E. Tajkhorshid et al., Scalable molecular dynamics with NAMD, J. Comput. Chem, vol.26, pp.1781-1802, 2005.

J. Huang, S. Rauscher, G. Nawrocki, T. Ran, M. Feig et al., CHARMM36m: An improved force field for folded and intrinsically disordered proteins, Nat. Methods, vol.14, pp.71-73, 2017.

V. Zoete, M. A. Cuendet, A. L. Grosdidier, and O. Michielin, SwissParam: A fast force field generation tool for small organic molecules, J. Comput. Chem, vol.32, pp.2359-2368, 2011.

P. Mark and L. Nilsson, Structure and dynamics of the TIP3P, SPC, and SPC/E water models at 298 K, J. Phys. Chem, vol.105, pp.9954-9960, 2001.

R. J. Shattock, B. F. Haynes, B. Pulendran, J. Flores, and J. Esparza, Improving defences at the portal of HIV entry: Mucosal and innate immunity, PLOS Med, vol.5, p.81, 2008.

J. A. Izaguirre, D. P. Catarello, J. M. Wozniak, and R. D. Skeel, Langevin stabilization of molecular dynamics, J. Chem. Phys, vol.114, pp.2090-2098, 2001.

A. E. Shaw, J. Hughes, Q. Gu, A. Behdenna, J. B. Singer et al., Fundamental properties of the mammalian innate immune system revealed by multispecies comparison of type I interferon responses, PLOS Biol, vol.15, p.2004086, 2017.

X. Xie, J. Lu, E. J. Kulbokas, T. R. Golub, V. Mootha et al., Systematic discovery of regulatory motifs in human promoters and 3? UTRs by comparison of several mammals, Nature, vol.434, pp.338-345, 2005.

A. Subramanian, P. Tamayo, V. K. Mootha, S. Mukherjee, B. L. Ebert et al., Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles, Proc. Natl. Acad. Sci. U.S.A, vol.102, pp.15545-15550, 2005.

S. Münch, D. Nisole, J. Duffy, and . Herbeuval,

S. Garcia, B. Charbit, N. Leboulanger, and B. Jahrsdörfer,

B. Bader-meunier, P. Quartier, C. Bodemer, V. Baudouin, Y. Dieudonné et al., , p.9019

, Sci Adv REFERENCES

, This article cites 70 articles, 20 of which you can access for free