T. Aki, T. Funakoshi, and K. Uemura, Regulated necrosis and its implications in toxicology, Toxicology, vol.333, pp.118-126, 2015.
DOI : 10.1016/j.tox.2015.04.003

S. Anguissola, D. Garry, A. Salvati, O. Brien, P. Dawson et al., High Content Analysis Provides Mechanistic Insights on the Pathways of Toxicity Induced by Amine-Modified Polystyrene Nanoparticles, PLoS ONE, vol.14, issue.(4), pp.108025-108035, 2014.
DOI : 10.1371/journal.pone.0108025.s001

, Phagocytes and oxidative stress, Babior BM Am J Med, vol.109, issue.100, pp.33-44, 2000.

S. Balasubramanian, K. Poh, C. Ong, W. Kreyling, W. Ong et al., The effect of primary particle size on biodistribution of inhaled gold nano-agglomerates, Biomaterials, vol.34, issue.22, pp.5439-5452, 2013.
DOI : 10.1016/j.biomaterials.2013.03.080

M. Bexiga, C. Kelly, K. Dawson, and J. Simpson, RNAi-mediated inhibition of apoptosis fails to prevent cationic nanoparticle-induced cell death in cultured cells, Nanomedicine, vol.9, issue.11, pp.1651-1664, 2014.
DOI : 10.1101/gad.1199904

M. Bexiga, J. Varela, and F. Wang, Cationic nanoparticles induce caspase 3-, 7- and 9-mediated cytotoxicity in a human astrocytoma cell line, Nanotoxicology, vol.270, issue.18, pp.557-567, 2011.
DOI : 10.1046/j.1432-1033.2003.03765.x

D. Bonvin, H. Hofmann, M. Ebersold, and M. , Assessment of nanoparticles' safety: corrected absorbance-based toxicity test doi:10.1039/c7an00382j Bradford MM (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding, Analyst Anal Biochem, vol.142, issue.72, pp.2338-2342248, 2017.

W. Chanput, J. Mes, and H. Wichers, THP-1 cell line: An in vitro cell model for immune modulation approach, International Immunopharmacology, vol.23, issue.1, pp.37-45, 2014.
DOI : 10.1016/j.intimp.2014.08.002

A. Coquette, N. Berna, A. Vandenbosch, M. Rosdy, D. Wever et al., Analysis of interleukin-1?? (IL-1??) and interleukin-8 (IL-8) expression and release in in vitro reconstructed human epidermis for the prediction of in vivo skin irritation and/or sensitization, Toxicology in Vitro, vol.17, issue.3, pp.311-332, 2003.
DOI : 10.1016/S0887-2333(03)00019-5

T. Cruz, R. Gaspar, A. Donato, and C. Lopes, Interaction between polyalkylcyanoacrylate nanoparticles and peritoneal macrophages: MTT metabolism, NBT reduction, and NO production, Pharmaceutical Research, vol.14, issue.1, pp.73-79, 1997.
DOI : 10.1023/A:1012059501947

J. Elliott, M. Rosslein, and N. Song, Toward achieving harmonization in a nanocytotoxicity assay measurement through an interlaboratory comparison study, ALTEX, vol.34, issue.2, pp.201-218, 2017.
DOI : 10.14573/altex.1605021

M. Esch, G. Mahler, T. Stokol, and M. Shuler, Body-on-a-chip simulation with gastrointestinal tract and liver tissues suggests that ingested nanoparticles have the potential to cause liver injury, Lab Chip, vol.32, issue.16, pp.3081-3092, 2014.
DOI : 10.1080/03639040500465991

H. Forman and M. Torres, Reactive Oxygen Species and Cell Signaling, American Journal of Respiratory and Critical Care Medicine, vol.166, issue.supplement_1, pp.4-8, 2002.
DOI : 10.1161/01.RES.83.7.730

K. Foster, C. Oster, M. Mayer, M. Avery, and K. Audus, Characterization of the A549 Cell Line as a Type II Pulmonary Epithelial Cell Model for Drug Metabolism, Experimental Cell Research, vol.243, issue.2, pp.359-366, 1998.
DOI : 10.1006/excr.1998.4172

L. Galluzzi, O. Kepp, S. Krautwald, G. Kroemer, and A. Linkermann, Molecular mechanisms of regulated necrosis, Seminars in Cell & Developmental Biology, vol.35, pp.24-32, 2014.
DOI : 10.1016/j.semcdb.2014.02.006

D. Grainger, Cell-based drug testing; this world is not flat, Advanced Drug Delivery Reviews, vol.69, issue.70, 2014.
DOI : 10.1016/j.addr.2014.04.001

D. Green, L. Galluzzi, and G. Kroemer, Cell biology Metabolic control of cell death, Science, vol.345, issue.6203, 2014.

A. C. , , p.22

C. Guguen-guillouzo, B. Clement, and G. Baffet, Maintenance and reversibility of active albumin secretion by adult rat hepatocytes co-cultured with another liver epithelial cell type, Experimental Cell Research, vol.143, issue.1, pp.47-54, 1983.
DOI : 10.1016/0014-4827(83)90107-6

T. Hartung, Towards a new toxicology ? evolution or revolution?, Altern. Lab. Anim, vol.36, pp.635-639, 2008.

M. Hermanns, R. Unger, K. Kehe, K. Peters, and C. Kirkpatrick, Lung epithelial cell lines in coculture with human pulmonary microvascular endothelial cells: development of an alveolo-capillary barrier in vitro, Laboratory Investigation, vol.286, issue.6 Spec No, pp.736-752, 2004.
DOI : 10.1152/ajplung.00187.2003

K. Hillgren, A. Kato, and R. Borchardt, In vitro systems for studying intestinal drug absorption, Medicinal Research Reviews, vol.71, issue.2, pp.83-109, 1995.
DOI : 10.1016/0016-5085(85)90646-8

M. Hjort, A. Brenyo, and J. Finkelstein, Alveolar epithelial cell-macrophage interactions affect oxygen-stimulated interleukin-8 release, Inflammation, vol.27, issue.3, pp.137-145, 2003.
DOI : 10.1023/A:1023817811850

R. Judson, R. Kavlock, M. Martin, D. Reif, K. Houck et al., Perspectives on validation of high-throughput assays supporting 21st century toxicity testing, ALTEX, vol.30, issue.1, pp.51-56, 2013.
DOI : 10.14573/altex.2013.1.051

C. Jumarie and C. Malo, Caco-2 cells cultured in serum-free medium as a model for the study of enterocytic differentiation in vitro, Journal of Cellular Physiology, vol.62, issue.1, pp.24-33, 1991.
DOI : 10.1111/j.1768-322X.1985.tb00399.x

L. Kangas, M. Gronroos, and A. Nieminen, Bioluminescence of cellular ATP: a new method for evaluating cytotoxic agents in vitro, Med Biol, vol.62, issue.6, pp.338-343, 1984.

M. Kendall and S. Holgate, Health impact and toxicological effects of nanomaterials in the lung, Respirology, vol.11, issue.1-3, pp.743-758, 2012.
DOI : 10.1007/s11051-008-9459-z

J. Kim, C. Aberg, G. De-carcer, M. Malumbres, A. Salvati et al., Low Dose of Amino-Modified Nanoparticles Induces Cell Cycle Arrest, ACS Nano, vol.7, issue.9, pp.7483-7494, 2013.
DOI : 10.1021/nn403126e

C. Korzeniewski and D. Callewaert, An enzyme-release assay for natural cytotoxicity, Journal of Immunological Methods, vol.64, issue.3, pp.313-320, 1983.
DOI : 10.1016/0022-1759(83)90438-6

W. Kreyling, S. Hirn, and W. Möller, Air???Blood Barrier Translocation of Tracheally Instilled Gold Nanoparticles Inversely Depends on Particle Size, ACS Nano, vol.8, issue.1, pp.222-233, 2014.
DOI : 10.1021/nn403256v

A. Kroll, M. Pillukat, D. Hahn, and J. Schnekenburger, Interference of engineered nanoparticles with in vitro toxicity assays, Archives of Toxicology, vol.25, issue.Suppl 1, pp.1123-1136, 2012.
DOI : 10.1016/j.dental.2009.03.012

H. Labouta and M. Schneider, Interaction of inorganic nanoparticles with the skin barrier: current status and critical review, Nanomedicine: Nanotechnology, Biology and Medicine, vol.9, issue.1, pp.39-54, 2013.
DOI : 10.1016/j.nano.2012.04.004

S. Lanone, F. Rogerieux, and J. Geys, Comparative toxicity of 24 manufactured nanoparticles in human alveolar epithelial and macrophage cell lines, Particle and Fibre Toxicology, vol.6, issue.1, pp.14-24, 2009.
DOI : 10.1186/1743-8977-6-14

URL : https://hal.archives-ouvertes.fr/inserm-00407214

J. Lewinson, W. Mayr, and H. Wagner, Characterization and Toxicological Behavior of Synthetic Amorphous Hydrophobic Silica, Regulatory Toxicology and Pharmacology, vol.20, issue.1, pp.37-57, 1994.
DOI : 10.1006/rtph.1994.1035

T. Loret, E. Peyret, and M. Dubreuil, Air-liquid interface exposure to aerosols of poorly soluble nanomaterials induces different biological activation levels compared to exposure to suspensions, Particle and Fibre Toxicology, vol.208, issue.3, pp.58-68, 2016.
DOI : 10.1186/1743-8977-11-5

URL : https://hal.archives-ouvertes.fr/ineris-01863163

K. Martin, The chemistry of silica and its potential health benefits, J Nutr Health Aging, vol.11, issue.2, pp.94-97, 2007.

Z. Mckenzie, M. Kendall, and R. Mackay, Surfactant protein A (SP-A) inhibits agglomeration and macrophage uptake of toxic amine modified nanoparticles, Nanotoxicology, vol.262, issue.8, pp.952-962, 2015.
DOI : 10.1080/08958370304478

A. C. , , p.23

N. Monteiro-riviere, A. Inman, and L. Zhang, Limitations and relative utility of screening assays to assess engineered nanoparticle toxicity in a human cell line, Toxicology and Applied Pharmacology, vol.234, issue.2, pp.222-235, 2009.
DOI : 10.1016/j.taap.2008.09.030

T. Mosmann, Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays, Journal of Immunological Methods, vol.65, issue.1-2, pp.55-63, 1983.
DOI : 10.1016/0022-1759(83)90303-4

K. Nam, A. Smith, S. Lone, S. Kwon, and D. Kim, Biomimetic 3D Tissue Models for Advanced High-Throughput Drug Screening, Journal of Laboratory Automation, vol.40, issue.3, pp.201-215, 2015.
DOI : 10.1098/rsif.2006.0179

I. Nelissen, I. Selderslaghs, R. Heuvel, H. Witters, G. Verheyen et al., MUTZ-3-derived dendritic cells as an in vitro alternative model to CD34+ progenitor-derived dendritic cells for testing of chemical sensitizers, Toxicology in Vitro, vol.23, issue.8, pp.1477-1481, 2009.
DOI : 10.1016/j.tiv.2009.08.022

G. Nohynek and E. Dufour, Nano-sized cosmetic formulations or solid nanoparticles in sunscreens: A risk to human health?, Archives of Toxicology, vol.13, issue.06, pp.1063-1075, 2012.
DOI : 10.1117/1.3041492

R. Peters, E. Kramer, and A. Oomen, Digestion of Foods Containing Silica as a Food Additive, ACS Nano, vol.6, issue.3, pp.2441-2451, 2012.
DOI : 10.1021/nn204728k

V. Petronilli, D. Penzo, L. Scorrano, P. Bernardi, D. Lisa et al., and Cell Death, Journal of Biological Chemistry, vol.256, issue.15, pp.12030-12034, 2001.
DOI : 10.1016/S0968-0004(00)01609-1

J. Piret, O. Bondarenko, and M. Boyles, Pan-European inter-laboratory studies on a panel of in vitro cytotoxicity and pro-inflammation assays for nanoparticles, Archives of Toxicology, vol.46, issue.3, pp.2315-2330, 2017.
DOI : 10.1021/ar300031y

J. Piret, J. Mejia, S. Lucas, C. Zouboulis, C. Saout et al., Sonicated and stirred copper oxide nanoparticles induce similar toxicity and pro-inflammatory response in N-hTERT keratinocytes and SZ95 sebocytes, Journal of Nanoparticle Research, vol.113, issue.6, pp.10-1007, 2014.
DOI : 10.1046/j.1523-1747.1999.00771.x

J. Piret, S. Vankoningsloo, and J. Mejia, Differential toxicity of copper (II) oxide nanoparticles of similar hydrodynamic diameter on human differentiated intestinal Caco-2 cell monolayers is correlated in part to copper release and shape, Nanotoxicology, vol.113, issue.6, pp.789-803, 2011.
DOI : 10.1046/j.1523-1747.1999.00771.x

Y. Poumay, F. Dupont, S. Marcoux, M. Leclercq-smekens, M. Herin et al., A simple reconstructed human epidermis: preparation of the culture model and utilization in in vitro studies, Archives of Dermatological Research, vol.121, issue.5, pp.203-214, 2004.
DOI : 10.1002/jcp.1041210106

B. Prietl, C. Meindl, E. Roblegg, T. Pieber, G. Lanzer et al., Nano-sized and micro-sized polystyrene particles affect phagocyte function, Proteomic and Lipidomic Analysis of Nanoparticle Corona upon Contact with Lung Surfactant Reveals Differences in Protein, but Not Lipid Composition, pp.1-16, 2014.
DOI : 10.1016/j.ejps.2010.09.006

M. Rösslein, J. Elliott, and M. Salit, Use of Cause-and-Effect Analysis to Design a High-Quality Nanocytotoxicology Assay, Chemical Research in Toxicology, vol.28, issue.1, pp.21-30, 2015.
DOI : 10.1021/tx500327y

P. Ruenraroengsak, P. Novak, and D. Berhanu, Respiratory epithelial cytotoxicity and membrane damage (holes) caused by amine-modified nanoparticles, Nanotoxicology, vol.42, issue.5, pp.94-108, 2011.
DOI : 10.1165/rcmb.2009-0138OC

J. Salomon, V. Muchitsch, and J. Gausterer, Model for Transport Studies of Human Distal Lung Epithelial Barrier, Molecular Pharmaceutics, vol.11, issue.3, pp.995-1006, 2014.
DOI : 10.1021/mp4006535

M. Skocaj, M. Filipic, J. Petkovic, and S. Novak, Titanium dioxide in our everyday life; is it safe?, Radiology and Oncology, vol.22, issue.4, pp.227-247, 2011.
DOI : 10.1016/0013-4686(93)80022-R

A. C. , , p.24

A. Stepanenko and V. Dmitrenko, Pitfalls of the MTT assay: Direct and off-target effects of inhibitors can result in over/underestimation of cell viability, Gene, vol.574, issue.2, pp.193-203, 2015.
DOI : 10.1016/j.gene.2015.08.009

L. Stoehr, C. Endes, and I. Radauer-preiml, Assessment of a panel of interleukin-8 reporter lung epithelial cell lines to monitor the pro-inflammatory response following zinc oxide nanoparticle exposure under different cell culture conditions, Particle and Fibre Toxicology, vol.86, issue.7, pp.29-39, 2015.
DOI : 10.1002/bit.20059

V. Stone, H. Johnston, and R. Schins, systems for nanotoxicology: methodological considerations, Critical Reviews in Toxicology, vol.105, issue.1, pp.613-626, 2009.
DOI : 10.1073/pnas.0805411105

J. Tournebize, A. Sapin-minet, G. Bartosz, P. Leroy, and A. Boudier, Pitfalls of assays devoted to evaluation of oxidative stress induced by inorganic nanoparticles, Talanta, vol.116, pp.753-763, 2013.
DOI : 10.1016/j.talanta.2013.07.077

URL : https://hal.archives-ouvertes.fr/hal-01272750

T. Tralau, M. Oelgeschlager, and R. Gurtler, Regulatory toxicology in the twenty-first century: challenges, perspectives and possible solutions, Archives of Toxicology, vol.42, issue.1, pp.823-850, 2015.
DOI : 10.1002/hast.104

S. Vankoningsloo, J. Piret, and C. Saout, Cytotoxicity of multi-walled carbon nanotubes in three skin cellular models: Effects of sonication, dispersive agents and corneous layer of reconstructed epidermis, Nanotoxicology, vol.113, issue.6, pp.84-97, 2010.
DOI : 10.1046/j.1523-1747.1999.00771.x

D. Volpe, Variability in Caco-2 and MDCK Cell-Based Intestinal Permeability Assays, Journal of Pharmaceutical Sciences, vol.97, issue.2, pp.712-725, 2008.
DOI : 10.1002/jps.21010

F. Wang, L. Yu, and M. Monopoli, The biomolecular corona is retained during nanoparticle uptake and protects the cells from the damage induced by cationic nanoparticles until degraded in the lysosomes, Nanomedicine: Nanotechnology, Biology and Medicine, vol.9, issue.8, pp.1159-1168, 2013.
DOI : 10.1016/j.nano.2013.04.010

A. Weir, P. Westerhoff, L. Fabricius, K. Hristovski, V. Goetz et al., Titanium Dioxide Nanoparticles in Food and Personal Care Products, Environmental Science & Technology, vol.46, issue.4, pp.2242-2250, 2012.
DOI : 10.1021/es204168d

URL : http://europepmc.org/articles/pmc3288463?pdf=render

R. Wottrich, S. Diabate, and H. Krug, Biological effects of ultrafine model particles in human macrophages and epithelial cells in mono- and co-culture, International Journal of Hygiene and Environmental Health, vol.207, issue.4, pp.353-361, 2004.
DOI : 10.1078/1438-4639-00300

T. Xia, R. Hamilton, and J. Bonner, Interlaboratory Evaluation of in Vitro Cytotoxicity and Inflammatory Responses to Engineered Nanomaterials: The NIEHS Nano GO Consortium, Environmental Health Perspectives, vol.121, issue.6, pp.683-690, 2013.
DOI : 10.1289/ehp.1306561

T. Xia, M. Kovochich, M. Liong, J. Zink, and A. Nel, Cationic Polystyrene Nanosphere Toxicity Depends on Cell-Specific Endocytic and Mitochondrial Injury Pathways, ACS Nano, vol.2, issue.1, pp.85-96, 2008.
DOI : 10.1021/nn700256c