J. Liu, D. A. Sonshine, S. Shervani, and R. H. Hurt, Controlled Release of Biologically Active Silver from Nanosilver Surfaces, ACS Nano, vol.4, issue.11, pp.6903-6913, 2010.

Z. M. Xiu, Q. B. Zhang, H. L. Puppala, V. L. Colvin, and P. J. Alvarez, Negligible Particle-Specific Antibacterial Activity of Silver Nanoparticles, Nano Letters, vol.12, issue.8, pp.4271-4275, 2012.

V. De-matteis, M. A. Malvindi, A. Galeone, V. Brunetti, E. De-luca et al., Negligible particle-specific toxicity mechanism of silver nanoparticles: The role of Ag+ ion release in the cytosol, Nanomedicine: Nanotechnology, Biology and Medicine, vol.11, issue.3, pp.731-739, 2015.

M. Marchioni, P. Jouneau, M. Chevallet, I. Michaud-soret, and A. Deniaud, Silver nanoparticle fate in mammals: Bridging in vitro and in vivo studies, Coordination Chemistry Reviews, vol.364, pp.118-136, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01792483

S. W. Wijnhoven, W. J. Peijnenburg, C. A. Herberts, W. I. Hagens, A. G. Oomen et al., Nano-silver ? a review of available data and knowledge gaps in human and environmental risk assessment, Nanotoxicology, vol.3, issue.2, pp.109-138, 2009.

K. Kulthong, S. Srisung, K. Boonpavanitchakul, W. Kangwansupamonkon, and R. Maniratanachote, Determination of silver nanoparticle release from antibacterial fabrics into artificial sweat, Particle and Fibre Toxicology, vol.7, issue.1, p.8, 2010.

. Maniratanachote, Determination of silver nanoparticle release from antibacterial fabrics into artificial sweat, Particle and fibre toxicology, 2008.

M. E. Quadros, R. T. Pierson, N. S. Tulve, R. Willis, K. Rogers et al., Release of Silver from Nanotechnology-Based Consumer Products for Children, Environmental Science & Technology, vol.47, issue.15, pp.8894-8901, 2013.

B. Reidy, A. Haase, A. Luch, K. A. Dawson, and I. Lynch, Mechanisms of Silver Nanoparticle Release, Transformation and Toxicity: A Critical Review of Current Knowledge and Recommendations for Future Studies and Applications, Materials, vol.6, issue.6, pp.2295-2350, 2013.

S. Agnihotri, S. Mukherji, and S. Mukherji, Immobilized silver nanoparticles enhance contact killing and show highest efficacy: elucidation of the mechanism of bactericidal action of silver, Nanoscale, vol.5, issue.16, p.7328, 2013.

K. Wu, Y. Yang, Y. Zhang, J. Deng, and C. Lin, Antimicrobial activity and cytocompatibility of silver nanoparticles coated catheters via a biomimetic surface functionalization strategy, International journal of nanomedicine, vol.10, pp.7241-7252, 2015.

S. Nakamura, M. Sato, Y. Sato, N. Ando, T. Takayama et al., Synthesis and Application of Silver Nanoparticles (Ag NPs) for the Prevention of Infection in Healthcare Workers, International Journal of Molecular Sciences, vol.20, issue.15, p.3620, 2019.

K. Park, E. Park, I. K. Chun, K. Choi, S. H. Lee et al., Bioavailability and Toxicokinetics of citrate-coated silver nanoparticles in rats, Archives of Pharmacal Research, vol.34, issue.1, pp.153-158, 2011.

P. Bove, M. A. Malvindi, S. S. Kote, R. Bertorelli, M. Summa et al., Dissolution test for risk assessment of nanoparticles: a pilot study, Nanoscale, vol.9, issue.19, pp.6315-6326, 2017.

E. C. Mimura, J. P. Favoreto, M. E. Favero, K. L. Bonifacio, T. S. Peixe et al., Silver serum levels in burned patients treated with silver sulfadiazine and its toxicity on inflammatory cells, Burns, vol.46, issue.5, pp.1120-1127, 2020.

M. Trop, M. Novak, S. Rodl, B. Hellbom, W. Kroell et al., Silver-Coated Dressing Acticoat Caused Raised Liver Enzymes and Argyria-like Symptoms in Burn Patient, The Journal of Trauma: Injury, Infection, and Critical Care, vol.60, issue.3, pp.648-652, 2006.

A. T. Wan, R. A. Conyers, C. J. Coombs, and J. P. Masterton, Determination of silver in blood, urine, and tissues of volunteers and burn patients, Clinical Chemistry, vol.37, issue.10, pp.1683-1687, 1991.

A. R. Gliga, S. Skoglund, I. Odnevall-wallinder, B. Fadeel, and H. L. Karlsson, Size-dependent cytotoxicity of silver nanoparticles in human lung cells: the role of cellular uptake, agglomeration and Ag release, Particle and Fibre Toxicology, vol.11, issue.1, p.11, 2014.

X. Jiang, T. Micl?u?, L. Wang, R. Foldbjerg, D. S. Sutherland et al., Fast intracellular dissolution and persistent cellular uptake of silver nanoparticles in CHO-K1 cells: implication for cytotoxicity, Nanotoxicology, vol.9, issue.2, pp.181-189, 2014.

R. P. Singh and P. Ramarao, Cellular uptake, intracellular trafficking and cytotoxicity of silver nanoparticles, Toxicology Letters, vol.213, issue.2, pp.249-259, 2012.

G. Veronesi, C. Aude-garcia, I. Kieffer, T. Gallon, P. Delangle et al., Exposure-dependent Ag+release from silver nanoparticles and its complexation in AgS2sites in primary murine macrophages, Nanoscale, vol.7, issue.16, pp.7323-7330, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01157525

G. Veronesi, A. Deniaud, T. Gallon, P. H. Jouneau, J. Villanova et al., Visualization, quantification and coordination of Ag+ions released from silver nanoparticles in hepatocytes, Nanoscale, vol.8, issue.38, pp.17012-17021, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01474979

L. Wang, T. Zhang, P. Li, W. Huang, J. Tang et al., Use of Synchrotron Radiation-Analytical Techniques To Reveal Chemical Origin of Silver-Nanoparticle Cytotoxicity, ACS Nano, vol.9, issue.6, pp.6532-6547, 2015.

N. W. Adams and J. R. Kramer, Potentiometric Determination of Silver Thiolate Formation Constants Using a Ag2S Electrode, Aqua. Geochem, vol.5, pp.1-11, 1999.

P. V. Asharani, G. Low-kah-mun, M. P. Hande, and S. Valiyaveettil, Cytotoxicity and Genotoxicity of Silver Nanoparticles in Human Cells, ACS Nano, vol.3, issue.2, pp.279-290, 2008.

H. R. Kim, M. J. Kim, S. Y. Lee, S. M. Oh, and K. H. Chung, Genotoxic effects of silver nanoparticles stimulated by oxidative stress in human normal bronchial epithelial (BEAS-2B) cells, Mutation Research/Genetic Toxicology and Environmental Mutagenesis, vol.726, issue.2, pp.129-135, 2011.

A. Azioune, N. Carpi, Q. Tseng, M. Théry, and M. Piel, Protein Micropatterns, Microtubules: in vivo, vol.97, pp.133-146, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00981469

A. Scherl, Y. Couté, C. Déon, A. Callé, K. Kindbeiter et al., Functional Proteomic Analysis of Human Nucleolus, Molecular Biology of the Cell, vol.13, issue.11, pp.4100-4109, 2002.
URL : https://hal.archives-ouvertes.fr/hal-00179628

C. R. Wooton-kee, A. K. Jain, M. Wagner, M. A. Grusak, M. J. Finegold et al., Elevated copper impairs hepatic nuclear receptor function in Wilson?s disease, Journal of Clinical Investigation, vol.125, issue.9, pp.3449-3460, 2015.

E. Kaneko, M. Matsuda, Y. Yamada, Y. Tachibana, I. Shimomura et al., Induction of Intestinal ATP-binding Cassette Transporters by a Phytosterol-derived Liver X Receptor Agonist, Journal of Biological Chemistry, vol.278, issue.38, pp.36091-36098, 2003.

G. Martínez-criado, J. Villanova, R. Tucoulou, D. Salomon, J. P. Suuronen et al., ID16B: a hard X-ray nanoprobe beamline at the ESRF for nano-analysis, Journal of Synchrotron Radiation, vol.23, issue.1, pp.344-352, 2016.

V. A. Solé, E. Papillon, M. Cotte, P. H. Walter, and J. Susini, A multiplatform code for the analysis of energy-dispersive X-ray fluorescence spectra, Spectrochimica Acta Part B: Atomic Spectroscopy, vol.62, issue.1, pp.63-68, 2007.

O. Mathon, A. Beteva, J. Borrel, D. Bugnazet, S. Gatla et al., The time-resolved and extreme conditions XAS (TEXAS) facility at the European Synchrotron Radiation Facility: the general-purpose EXAFS bending-magnet beamline BM23, Journal of Synchrotron Radiation, vol.22, issue.6, pp.1548-1554, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01573036

B. Ravel and M. Newville, ATHENA,ARTEMIS,HEPHAESTUS: data analysis for X-ray absorption spectroscopy usingIFEFFIT, Journal of Synchrotron Radiation, vol.12, issue.4, pp.537-541, 2005.

J. Schindelin, I. Arganda-carreras, E. Frise, V. Kaynig, M. Longair et al., Fiji: an open-source platform for biological-image analysis, Nature Methods, vol.9, issue.7, pp.676-682, 2012.
URL : https://hal.archives-ouvertes.fr/pasteur-02616466

C. Sommer, C. Straehle, U. Kothe, and F. A. Hamprecht, Ilastik: Interactive learning and segmentation toolkit, 2011 IEEE International Symposium on Biomedical Imaging: From Nano to Macro, 2011.

M. Chevallet, B. Gallet, A. Fuchs, P. H. Jouneau, K. Um et al., Metal homeostasis disruption and mitochondrial dysfunction in hepatocytes exposed to sub-toxic doses of zinc oxide nanoparticles, Nanoscale, vol.8, issue.43, pp.18495-18506, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01474070

M. Cuillel, M. Chevallet, P. Charbonnier, C. Fauquant, I. Pignot-paintrand et al., Interference of CuO nanoparticles with metal homeostasis in hepatocytes under sub-toxic conditions, Nanoscale, vol.6, issue.3, pp.1707-1715, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00936382

A. P. Gondikas, A. Morris, B. C. Reinsch, S. M. Marinakos, G. V. Lowry et al., Cysteine-Induced Modifications of Zero-valent Silver Nanomaterials: Implications for Particle Surface Chemistry, Aggregation, Dissolution, and Silver Speciation, Environmental Science & Technology, vol.46, issue.13, pp.7037-7045, 2012.

M. Marchioni, T. Gallon, I. Worms, P. Jouneau, C. Lebrun et al., Insights into polythiol-assisted AgNP dissolution induced by bio-relevant molecules, Environmental Science: Nano, vol.5, issue.8, pp.1911-1920, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01869612

J. Lee, M. M. Peña, Y. Nose, and D. J. Thiele, Biochemical Characterization of the Human Copper Transporter Ctr1, Journal of Biological Chemistry, vol.277, issue.6, pp.4380-4387, 2001.

K. I. Farley, Y. Surovtseva, J. Merkel, and S. J. Baserga, Determinants of mammalian nucleolar architecture, Chromosoma, vol.124, issue.3, pp.323-331, 2015.

C. Goodpasture and S. E. Bloom, Visualization of nucleolar organizer regions in mammalian chromosomes using silver staining, Chromosoma, vol.53, issue.1, pp.37-50, 1975.

P. Roussel and D. Hernandez-verdun, Identification of Ag-NOR Proteins, Markers of Proliferation Related to Ribosomal Gene Activity, Experimental Cell Research, vol.214, issue.2, pp.465-472, 1994.

P. Roussel, P. Belenguer, F. Amalric, and D. Hernandez-verdun, Nucleolin is an Ag-NOR protein; This property is determined by its amino-terminal domain independently of its phosphorylation state, Experimental Cell Research, vol.203, issue.1, pp.259-269, 1992.

J. L. Woolford and S. J. Baserga, Ribosome Biogenesis in the YeastSaccharomyces cerevisiae, Genetics, vol.195, issue.3, pp.643-681, 2013.

C. E. Foulds, L. S. Treviño, B. York, and C. L. Walker, Endocrine-disrupting chemicals and fatty liver disease, Nature Reviews Endocrinology, vol.13, issue.8, pp.445-457, 2017.

B. J. Deegan, A. M. Bona, V. Bhat, D. C. Mikles, C. B. Mcdonald et al., Structural and thermodynamic consequences of the replacement of zinc with environmental metals on estrogen receptor ?-DNA interactions, Journal of Molecular Recognition, vol.24, issue.6, pp.1007-1017, 2011.

T. W. Hutchens, M. H. Allen, C. M. Li, and T. Yip, Occupancy of a C2 -C2 type ?Zinc-finger? protein domain by copper Direct observation by electrospray ionization mass spectrometry, FEBS Letters, vol.309, issue.2, pp.170-174, 1992.

P. F. Predki and B. Sarkar, Effect of replacement of ?zinc finger? zinc on the DNA binding properties of the estrogen receptor, Journal of Inorganic Biochemistry, vol.43, issue.2-3, p.511, 1991.

J. P. Hamilton, L. Koganti, A. Muchenditsi, V. S. Pendyala, D. Huso et al., Activation of liver X receptor/retinoid X receptor pathway ameliorates liver disease inAtp7B?/?(Wilson disease) mice, Hepatology, vol.63, issue.6, pp.1828-1841, 2016.

P. A. Wilmarth, K. K. Short, O. Fiehn, S. Lutsenko, L. L. David et al., A systems approach implicates nuclear receptor targeting in the Atp7b?/? mouse model of Wilson's disease, Metallomics, vol.4, issue.7, p.660, 2012.

C. S. Kim, X. Li, Y. Jiang, B. Yan, G. Y. Tonga et al., Cellular imaging of endosome entrapped small gold nanoparticles, MethodsX, vol.2, pp.306-315, 2015.

K. Narayan and S. Subramaniam, Focused ion beams in biology, Nature Methods, vol.12, issue.11, pp.1021-1031, 2015.

K. Wrzesinski, M. C. Magnone, L. V. Hansen, M. E. Kruse, T. Bergauer et al., HepG2/C3A 3D spheroids exhibit stable physiological functionality for at least 24 days after recovering from trypsinisation, Toxicology Research, vol.2, issue.3, p.163, 2013.

D. E. Johnson, P. Ostrowski, V. Jaumouillé, and S. Grinstein, The position of lysosomes within the cell determines their luminal pH, Journal of Cell Biology, vol.212, issue.6, pp.677-692, 2016.

B. E. Black, J. M. Holaska, F. Rastinejad, and B. M. Paschal, DNA binding domains in diverse nuclear receptors function as nuclear export signals, Current Biology, vol.11, issue.22, pp.1749-1758, 2001.

M. S. Mavinakere, J. M. Powers, K. S. Subramanian, V. R. Roggero, and L. A. Allison, Multiple Novel Signals Mediate Thyroid Hormone Receptor Nuclear Import and Export, Journal of Biological Chemistry, vol.287, issue.37, pp.31280-31297, 2012.

A. Helleboid-chapman, S. Helleboid, H. Jakel, C. Timmerman, C. Sergheraert et al., Glucose regulates LXR? subcellular localization and function in rat pancreatic ?-cells, Cell Research, vol.16, issue.7, pp.661-670, 2006.

K. Kluska, M. D. Peris-díaz, D. P?onka, A. Moysa, M. Dadlez et al., Formation of highly stable multinuclear AgnSn clusters in zinc fingers disrupts their structure and function, Chemical Communications, vol.56, issue.9, pp.1329-1332, 2020.
URL : https://hal.archives-ouvertes.fr/hal-02494376

. Krezel, Formation of highly stable multinuclear AgnSn clusters in zinc fingers disrupts their structure and function, Chemical communications, vol.56, pp.1329-1332, 2020.
URL : https://hal.archives-ouvertes.fr/hal-02494376

A. Klug, The Discovery of Zinc Fingers and Their Applications in Gene Regulation and Genome Manipulation, Annual Review of Biochemistry, vol.79, issue.1, pp.213-231, 2010.

M. Marchioni, G. Veronesi, I. Worms, W. L. Ling, T. Gallon et al., Safer-by-design biocides made of tri-thiol bridged silver nanoparticle assemblies, Nanoscale Horizons, vol.5, issue.3, pp.507-513, 2020.
URL : https://hal.archives-ouvertes.fr/hal-02366178

Y. G. Yuan, Q. L. Peng, and S. Gurunathan, Silver nanoparticles enhance the apoptotic potential of gemcitabine in human ovarian cancer cells: combination therapy for effective cancer treatment, International Journal of Nanomedicine, vol.Volume 12, pp.6487-6502, 2017.

F. Peng, M. I. Setyawati, J. K. Tee, X. Ding, J. Wang et al., Nanoparticles promote in vivo breast cancer cell intravasation and extravasation by inducing endothelial leakiness, Nature Nanotechnology, vol.14, issue.3, pp.279-286, 2019.