, « glutathione metabolism » comprenant notamment les protéines Gpx1, Gss, Gstm2, Gstm4, Lap3 ; « regulation of actin cytoskeleton

. Actn4, Nous avons également observé la présence d'un réseau d'interaction comprenant diverses histones (H1, H2 et H3) comme mentionné ci-dessus, Un réseau central est constitué de protéines impliquées dans la voie « Glucose metabolism » notamment Eno2, Pgam1, Pkm, Pygl, Txnrd2, ainsi que dans le processus biologique « oxidation-reduction » comme Gpx1, Mdh2, Prdx2, Prdx5, Pygl, Txnrd1 et Txnrd2. De manière intéressante, ces 7 réseaux interagissent directement les uns avec les autres, ou indirectement via d'autres protéines

K. Adachi, N. Yamada, K. Yamamoto, Y. Y. Yamamoto, and O. , In vivo effect of industrial titanium dioxide nanoparticles experimentally exposed to hairless rat skin, Nanotoxicology, vol.4, pp.296-306, 2010.

. Afssaps, Nanoparticules de dioxyde de titane et d'oxyde de zinc dans les produits cosmétiques : Etat des connaissances sur la pénétration cutanée, génotoxicité et cancérogenèse -Point d'information, Journal, 2011.

. Afssaps, Recommandations relatives à l'utilisation des nanoparticules de dioxyde de titane et d'oxyde de zinc en tant que filtres ultraviolets dans les produits cosmétiques, Journal, 2011.

N. R. Aggarwal, K. Ls, and D. Fr, Diverse macrophage populations mediate acute lung inflammation and resolution, American Journal of Physiology-Lung Cellular and Molecular Physiology, vol.306, pp.709-725, 2014.

L. Ahlinder, B. Ekstrand-hammarstrom, G. P. Osterlund, and L. , Large uptake of titania and iron oxide nanoparticles in the nucleus of lung epithelial cells as measured by Raman imaging and multivariate classification, Biophysical journal, vol.105, pp.310-319, 2013.

A. Asgharian and B. , A multiple-path model of particle deposition in the rat lung, Fundamental and applied toxicology : official journal of the Society of Toxicology, vol.28, pp.41-50, 1995.

. Anses, Evaluation des risques liés aux nanomatériaux, enjeux et mise à jour des connaissances, Journal, 2014.

. Anses, Évaluation des risques liés aux nanomatériaux pour la population générale, les travailleurs et pour l'environnement, Journal, 2016.

. Anses, Dioxyde de titane, Journal, 2017.

. Anses, Nanoparticules de dioxyde de titane dans l'alimentation (additif E 171) : des effets biologiques qui doivent être confirmés, Journal, 2017.

L. Armand, M. Biola-clier, L. Bobyk, V. Collin-faure, H. Diemer et al., Molecular responses of alveolar epithelial A549 cells to chronic exposure to titanium dioxide nanoparticles: A proteomic view, Journal of proteomics, vol.134, pp.163-173, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01291979

L. Armand, A. Tarantini, D. Beal, M. Biola-clier, L. Bobyk et al., Long-term exposure of A549 cells to titanium dioxide nanoparticles induces DNA damage and sensitizes cells towards genotoxic agents, Nanotoxicology, vol.10, pp.913-923, 2016.
URL : https://hal.archives-ouvertes.fr/cea-01651136

R. A. Baan, Carcinogenic hazards from inhaled carbon black, titanium dioxide, and talc not containing asbestos or asbestiform fibers: recent evaluations by an IARC Monographs Working Group, Inhalation toxicology, vol.19, pp.213-228, 2007.

A. M. Bannunah, D. Vllasaliu, J. Lord, and S. Stolnik, Mechanisms of nanoparticle internalization and transport across an intestinal epithelial cell model: effect of size and surface charge, Molecular pharmaceutics, vol.11, pp.4363-4373, 2014.

M. Bartneck, H. A. Keul, . Zwadlo-klarwasser-g, and J. Groll, Phagocytosis independent extracellular nanoparticle clearance by human immune cells, Nano letters, vol.10, pp.59-63, 2010.

E. Bermudez, J. B. Mangum, B. Asgharian, B. A. Wong, E. E. Reverdy et al.,

, Long-term pulmonary responses of three laboratory rodent species to subchronic inhalation of pigmentary titanium dioxide particles, Toxicological sciences : an official journal of the Society of Toxicology, vol.70, pp.86-97

E. Bermudez, J. B. Mangum, B. A. Wong, B. Asgharian, P. M. Hext et al., Pulmonary responses of mice, rats, and hamsters to subchronic inhalation of ultrafine titanium dioxide particles, Toxicological sciences : an official journal of the Society of Toxicology, vol.77, pp.347-357, 2004.

D. M. Bernstein, Synthetic vitreous fibers: a review toxicology, epidemiology and regulations, Critical reviews in toxicology, vol.37, pp.839-886, 2007.

S. Bettini, E. Boutet-robinet, C. Cartier, C. Comera, E. Gaultier et al., Food-grade TiO2 impairs intestinal and systemic immune homeostasis, initiates preneoplastic lesions and promotes aberrant crypt development in the rat colon, Scientific reports, vol.7, p.40373, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01508951

S. Binet, M. Ricaud, S. Chazelet, J. Fontaine, L. Gaté et al., Dioxyde de titane nanométrique : de la nécessité de proposer une valeur limite d'exposition professionnelle. INRS, 17. Biswas SK. 2016. Does the interdependence between oxidative stress and inflammation explain the antioxidant paradox? Oxidative medicine and cellular longevity, 2016.

P. Boffetta, V. Gaborieau, L. Nadon, M. F. Parent, W. E. Siemiatycki et al., Exposure to titanium dioxide and risk of lung cancer in a population-based study from Montreal. Scandinavian journal of work, environment & health, vol.27, pp.227-232, 2001.

A. Boitelle, Mécanismes de recrutement et de régulation de l'activité des macrophages alvéolaires, 1997.

P. Borm, C. Fr, and G. Oberdorster, Lung particle overload: old school -new insights?, Particle and fibre toxicology, vol.12, p.10, 2015.

P. J. Borm, D. Hohr, Y. Steinfartz, Z. I. , and A. C. , Chronic Inflammation and Tumor Formation in Rats After Intratracheal Instillation of High Doses of Coal Dusts, Titanium Dioxides, and Quartz, Inhalation toxicology, vol.12, pp.225-231, 2000.

J. A. Bourdon, S. Halappanavar, A. T. Saber, N. R. Jacobsen, A. Williams et al., Hepatic and pulmonary toxicogenomic profiles in mice intratracheally instilled with carbon black nanoparticles reveal pulmonary inflammation, acute phase response, and alterations in lipid homeostasis, Toxicological sciences : an official journal of the Society of Toxicology, vol.127, pp.474-484, 2012.

J. A. Bourdon, A. Williams, B. Kuo, I. Moffat, P. A. White et al., Gene expression profiling to identify potentially relevant disease outcomes and support human health risk assessment for carbon black nanoparticle exposure, Toxicology, vol.303, pp.83-93, 2013.

A. Brazma, P. Hingamp, J. Quackenbush, G. Sherlock, P. Spellman et al., Minimum information about a microarray experiment (MIAME)-toward standards for microarray data, Nature genetics, vol.29, pp.365-371, 2001.

D. Brosson, Proteomic analysis and characterization of new spore wall proteins of the microsporidia Encephalitozoon cuniculi, an emerging pathogen of humans, 2006.
URL : https://hal.archives-ouvertes.fr/tel-00688692

C. L. Browning, T. The, M. Md, J. P. Wise, and . Sr, Titanium Dioxide Nanoparticles are not Cytotoxic or Clastogenic in Human Skin Cells, Journal of environmental & analytical toxicology, vol.4, 2014.

E. Brun, M. Carriere, and A. Mabondzo, In vitro evidence of dysregulation of blood-brain barrier function after acute and repeated/long-term exposure to TiO(2) nanoparticles, Biomaterials, vol.33, pp.886-896, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01930653

S. Brunauer, E. Ph, and E. Teller, Adsorption of Gases in Multimolecular Layers, Journal of the American Chemical Society, vol.60, pp.309-319, 1938.

L. Cabon, A. C. Martinez-torres, and S. A. Susin, Medecine sciences : M/S, vol.29, pp.1117-1124, 2013.

. Cancer-environnement, Le dioxyde de titane (TiO2), Journal, 2018.

Y. Cao, Y. Gong, W. Liao, Y. Luo, C. Wu et al., A review of cardiovascular toxicity of TiO 2, ZnO and Ag nanoparticles (NPs), pp.1-20, 2018.

M. H. Cha, T. Rhim, K. H. Kim, A. S. Jang, P. Yk et al., Proteomic identification of macrophage migrationinhibitory factor upon exposure to TiO2 particles, Molecular & cellular proteomics : MCP, vol.6, pp.56-63, 2007.

S. Chantepie, S. Krieger, and S. Leporrier, Méthylation dans les hémopathies malignes : techniques et perspectives, 2012.

, Hématologie, vol.18, pp.124-132

S. Charles, S. Jomini, V. Fessard, E. Bigorgne-vizade, R. C. Michel et al., Assessment of the in vitro genotoxicity of TiO2 nanoparticles in a regulatory context, Nanotoxicology, vol.12, pp.357-374, 2018.
URL : https://hal.archives-ouvertes.fr/anses-01741160

Q. Chaudhry, Opinion of the Scientific Committee on Consumer safety (SCCS) -Revision of the opinion on the safety of the use of titanium dioxide, nano form, in cosmetic products, Regulatory toxicology and pharmacology : RTP, vol.73, pp.669-670, 2015.

C. Jl and W. E. Fayerweather, Epidemiologic study of workers exposed to titanium dioxide, Journal of occupational medicine : official publication of the Industrial Medical Association, vol.30, pp.937-942, 1988.

M. Chen, M. Zhang, B. J. Tong, and W. , A decade of toxicogenomic research and its contribution to toxicological science, Toxicological sciences : an official journal of the Society of Toxicology, vol.130, pp.217-228, 2012.

Q. Chen, N. Wang, M. Zhu, J. Lu, H. Zhong et al., TiO2 nanoparticles cause mitochondrial dysfunction, activate inflammatory responses, and attenuate phagocytosis in macrophages: A proteomic and metabolomic insight, Redox biology, vol.15, pp.266-276, 2018.
DOI : 10.1016/j.redox.2017.12.011

URL : https://doi.org/10.1016/j.redox.2017.12.011

S. Chen, R. Yin, K. Mutze, Y. Yu, S. Takenaka et al., No involvement of alveolar macrophages in the initiation of carbon nanoparticle induced acute lung inflammation in mice, Particle and fibre toxicology, vol.13, p.33, 2016.

T. Chen, Y. J. Li, and Y. , Genotoxicity of titanium dioxide nanoparticles, Journal of food and drug analysis, vol.22, pp.95-104, 2014.
DOI : 10.1016/j.jfda.2014.01.008

URL : https://doi.org/10.1016/j.jfda.2014.01.008

Z. Chen, Y. Wang, T. Ba, Y. Li, J. Pu et al., Genotoxic evaluation of titanium dioxide nanoparticles in vivo and in vitro, Toxicology letters, vol.226, pp.314-319, 2014.

Z. Chen, Y. Wang, X. Wang, L. Zhuo, S. Chen et al., Effect of titanium dioxide nanoparticles on glucose homeostasis after oral administration, Journal of applied toxicology : JAT, vol.38, pp.810-823, 2018.

Z. Chen, Y. Wang, L. Zhuo, S. Chen, L. Zhao et al., Interaction of titanium dioxide nanoparticles with glucose on young rats after oral administration, Nanomedicine : nanotechnology, biology, and medicine, vol.11, pp.1633-1642, 2015.
DOI : 10.1016/j.nano.2015.06.002

Z. Chen, Y. Wang, L. Zhuo, S. Chen, L. Zhao et al., Effect of titanium dioxide nanoparticles on the cardiovascular system after oral administration, Toxicology letters, vol.239, pp.123-130, 2015.

D. B. Chithrani, Intracellular uptake, transport, and processing of gold nanostructures, Molecular membrane biology, vol.27, pp.299-311, 2010.

W. S. Cho, B. C. Kang, J. K. Lee, J. Jeong, C. J. Seok et al., Comparative absorption, distribution, and excretion of titanium dioxide and zinc oxide nanoparticles after repeated oral administration, Particle and fibre toxicology, vol.10, p.9, 2013.
DOI : 10.1186/1743-8977-10-9

URL : https://particleandfibretoxicology.biomedcentral.com/track/pdf/10.1186/1743-8977-10-9

C. , Fiche d'information sur la définition des nanomatériaux, 2014.

P. Collas, The current state of chromatin immunoprecipitation, Molecular biotechnology, vol.45, pp.87-100, 2010.

C. Williams and T. J. , Eotaxin and the attraction of eosinophils to the asthmatic lung, Respiratory research, vol.2, pp.150-156, 2001.

C. Pagnoni and A. , TiO2 in commercial sunscreen lotion: flow field-flow fractionation and ICP-AES together for size analysis, Analytical chemistry, vol.80, pp.7594-7608, 2008.

F. Cosnier, S. Bau, S. Grossmann, H. Nunge, C. Brochard et al., Design and Characterization of an Inhalation System to Expose Rodents to, Nanoaerosols. Aerosol and Air Quality Research, 2016.
DOI : 10.4209/aaqr.2016.01.0034

C. Pm and B. Fadeel, Emerging systems biology approaches in nanotoxicology: Towards a mechanism-based understanding of nanomaterial hazard and risk, Toxicology and applied pharmacology, vol.299, pp.101-111, 2016.

O. Creutzenberg, B. Bellmann, R. Korolewitz, W. Koch, I. Mangelsdorf et al., Change in agglomeration status and toxicokinetic fate of various nanoparticles in vivo following lung exposure in rats, Inhalation toxicology, vol.24, pp.821-830, 2012.

R. T. Cullen, C. L. Tran, D. Buchanan, J. M. Davis, A. Searl et al., Inhalation of poorly soluble particles. I. Differences in inflammatory response and clearance during exposure, Inhalation toxicology, vol.12, pp.1089-1111, 2000.

C. Bertke and S. , Exposure characterization of metal oxide nanoparticles in the workplace, Journal of occupational and environmental hygiene, vol.8, pp.580-587, 2011.

K. Daigo, Y. Takamatsu, and T. Hamakubo, The Protective Effect against Extracellular Histones Afforded by LongPentraxin PTX3 as a Regulator of NETs, Frontiers in immunology, vol.7, p.344, 2016.

D. Dankovic, K. E. Wheeler, and M. , An approach to risk assessment for TiO2, Inhalation toxicology, vol.19, pp.205-212, 2007.

D. Kastner and P. , Fold change rank ordering statistics: a new method for detecting differentially expressed genes, BMC bioinformatics, vol.15, p.14, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-00935627

S. L. Deshmane, S. Kremlev, S. Amini, and B. E. Sawaya, Monocyte chemoattractant protein-1 (MCP-1): an overview, Journal of interferon & cytokine research, vol.29, pp.313-326, 2009.

C. Detaille, Pathologie respiratoire des rongeurs et lagomorphes de compagnie, 2008.

C. A. Dick, D. M. Brown, D. K. Stone, and V. , The role of free radicals in the toxic and inflammatory effects of four different ultrafine particle types, Inhalation toxicology, vol.15, pp.39-52, 2003.

C. Disdier, J. Devoy, A. Cosnefroy, M. Chalansonnet, N. Herlin-boime et al., Tissue biodistribution of intravenously administrated titanium dioxide nanoparticles revealed blood-brain barrier clearance and brain inflammation in rat, Particle and fibre toxicology, vol.12, p.27, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01259856

K. Donaldson, P. J. Borm, G. Oberdorster, K. E. Pinkerton, V. Stone et al., Concordance between in vitro and in vivo dosimetry in the proinflammatory effects of low-toxicity, low-solubility particles: the key role of the proximal alveolar region, Inhalation toxicology, vol.20, pp.53-62, 2008.

K. Donaldson, L. Xy, and W. Macnee, Ultrafine (nanometre) particle mediated lung injury, J Aerosol Sci, vol.29, pp.553-560, 1998.

K. E. Driscoll, D. L. Costa, G. Hatch, R. Henderson, G. Oberdorster et al., Intratracheal instillation as an exposure technique for the evaluation of respiratory tract toxicity: uses and limitations, Toxicological sciences : an official journal of the Society of Toxicology, vol.55, pp.24-35, 2000.

K. E. Driscoll, L. C. Deyo, J. M. Carter, B. W. Howard, H. Dg et al., Effects of particle exposure and particle-elicited inflammatory cells on mutation in rat alveolar epithelial cells, Carcinogenesis, vol.18, pp.423-430, 1997.

. Ecetoc, TR 122 -Poorly Soluble Particles / Lung Overload. Brussels: European Centre for Ecotoxicology and Toxicology of Chemicals, 2013.

E. Barrett and T. , NCBI GEO standards and services for microarray data, Nature biotechnology, vol.24, pp.1471-1472, 2006.

N. El-sharkawy, S. Hamza, and E. Abou-zeid, Toxic impact of titanium dioxide (TiO2) in male albino rats with special reference to its effect on reproductive system, J Am Sci, vol.6, pp.865-872, 2010.

E. B. Engler-chiurazzi, P. A. Stapleton, J. J. Stalnaker, X. Ren, H. Hu et al., Impacts of prenatal nanomaterial exposure on male adult Sprague-Dawley rat behavior and cognition, Journal of toxicology and environmental health Part A, vol.79, pp.447-452, 2016.

. Eu, DIRECTIVE 2010/63/EU OF THE EUROPEAN PARLIAMENT AND OF THE COUNCIL of, p.22, 2010.

, on the protection of animals used for scientific purposes, 2010.

C. Fan, L. W. Meuchel, Q. Su, D. J. Angelini, A. Zhang et al., Resistin-Like Molecule alpha in Allergen-Induced Pulmonary Vascular Remodeling, American journal of respiratory cell and molecular biology, vol.53, pp.303-313, 2015.

J. Ferin, E. Oberdorster, S. Sc, and R. Gelein, Pulmonary Tissue Access of Ultrafine Particles, Journal of Aerosol Medicine, vol.4, pp.57-68, 1991.

J. Ferin, G. Oberdorster, and D. P. Penney, Pulmonary retention of ultrafine and fine particles in rats, American journal of respiratory cell and molecular biology, vol.6, pp.535-542, 1992.

A. T. Florence, Nanoparticle uptake by the oral route: Fulfilling its potential?, Drug discovery today Technologies, vol.2, pp.75-81, 2005.

G. Folkerts, J. Kloek, M. Rb, and F. P. Nijkamp, Reactive nitrogen and oxygen species in airway inflammation, European journal of pharmacology, vol.429, pp.251-262, 2001.

T. Fournier, M. Nn, and D. Porquet, Alpha-1-acid glycoprotein, Biochimica et biophysica acta, vol.1482, pp.157-171, 2000.

E. Froehlich and S. Salar-behzadi, Toxicological Assessment of Inhaled Nanoparticles: Role of in Vivo, ex Vivo, Silico Studies, 2014.

J. P. Fryzek, B. Chadda, D. Marano, K. White, S. Schweitzer et al., A cohort mortality study among titanium dioxide manufacturing workers in the United States, Journal of occupational and environmental medicine, vol.45, pp.400-409, 2003.

G. Fuchs, C. Diges, L. A. Kohlstaedt, K. A. Wehner, and P. Sarnow, Proteomic analysis of ribosomes: translational control of mRNA populations by glycogen synthase GYS1, Journal of molecular biology, vol.410, pp.118-130, 2011.

N. Fujiwara and K. Kobayashi, Macrophages in inflammation, Current drug targets Inflammation and allergy, vol.4, pp.281-286, 2005.

H. Gao, X. He, M. Wu, Z. Zhang, D. Wang et al., Proteomic analysis of differentially expressed proteins between stenotic and normal colon segment tissues derived from patients with Hirschsprung's disease. The protein journal, vol.30, pp.138-142, 2011.

Y. Gao, N. V. Gopee, P. C. Howard, and L. R. Yu, Proteomic analysis of early response lymph node proteins in mice treated with titanium dioxide nanoparticles, Journal of proteomics, vol.74, pp.2745-2759, 2011.

D. H. Garabrant, L. J. Fine, C. Oliver, L. Bernstein, and J. M. Peters, Abnormalities of pulmonary function and pleural disease among titanium metal production workers, Scandinavian journal of work, vol.13, pp.47-51, 1987.

L. Gate, C. Disdier, F. Cosnier, F. Gagnaire, J. Devoy et al., , 2017.

, Biopersistence and translocation to extrapulmonary organs of titanium dioxide nanoparticles after subacute inhalation exposure to aerosol in adult and elderly rats, Toxicology letters, vol.265, pp.61-69

Y. Ge, M. Bruno, K. Wallace, W. Winnik, and R. Y. Prasad, Proteome profiling reveals potential toxicity and detoxification pathways following exposure of BEAS-2B cells to engineered nanoparticle titanium dioxide, Proteomics, vol.11, pp.2406-2422, 2011.

M. Geiser, Update on macrophage clearance of inhaled micro-and nanoparticles, Journal of aerosol medicine and pulmonary drug delivery, vol.23, pp.207-217, 2010.

M. Geiser and W. G. Kreyling, Deposition and biokinetics of inhaled nanoparticles, Particle and fibre toxicology, vol.7, issue.2, 2010.

M. Geiser, B. Rothen-rutishauser, N. Kapp, S. Schurch, W. Kreyling et al., Ultrafine particles cross cellular membranes by nonphagocytic mechanisms in lungs and in cultured cells, Environmental health perspectives, vol.113, pp.1555-1560, 2005.

I. George, G. Naudin, S. Boland, S. Mornet, V. Contremoulins et al., Metallic oxide nanoparticle translocation across the human bronchial epithelial barrier, Nanoscale, vol.7, pp.4529-4544, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01228101

L. Geraets, A. G. Oomen, P. Krystek, N. R. Jacobsen, H. Wallin et al.,

, Tissue distribution and elimination after oral and intravenous administration of different titanium dioxide nanoparticles in rats, Particle and fibre toxicology, vol.11, p.30

J. Geys, A. Nemmar, E. Verbeken, E. Smolders, M. Ratoi et al., Acute toxicity and prothrombotic effects of quantum dots: impact of surface charge, Environmental health perspectives, vol.116, p.1607, 2008.

G. Martinez and F. O. , Alternative activation of macrophages: mechanism and functions, Immunity, vol.32, pp.593-604, 2010.

. Grailer and P. A. Ward, Lung inflammation and damage induced by extracellular histones, Inflammation and cell, vol.1, 2014.

V. H. Grassian, . O'shaughnessy-p-t, A. Adamcakova-dodd, P. Jm, and P. S. Thorne, Inhalation exposure study of titanium dioxide nanoparticles with a primary particle size of 2 to 5 nm, Environmental health perspectives, vol.115, pp.397-402, 2007.

H. Greim, P. Borm, R. Schins, K. Donaldson, K. Driscoll et al., TOXICITY OF FIBERS AND PARTICLES? REPORT OF THE WORKSHOP HELD, vol.13, pp.737-754, 2000.

L. Griffis, R. Wolff, R. Beethe, C. Hobbs, and R. Mcclellan, Pulmonary deposition of a/sup 99m/Tc-labeled aerosol in a whole-body exposure, 1979.

Y. Guichard, J. Schmit, C. Darne, L. Gate, M. Goutet et al., Cytotoxicity and genotoxicity of nanosized and microsized titanium dioxide and iron oxide particles in Syrian hamster embryo cells, The Annals of occupational hygiene, vol.56, pp.631-644, 2012.

N. L. Guo, Y. W. Wan, J. Denvir, D. W. Porter, M. Pacurari et al., Multiwalled carbon nanotube-induced gene signatures in the mouse lung: potential predictive value for human lung cancer risk and prognosis, Journal of toxicology and environmental health Part A, vol.75, pp.1129-1153, 2012.

J. R. Gurr, A. S. Wang, C. H. Chen, and J. Ky, Ultrafine titanium dioxide particles in the absence of photoactivation can induce oxidative damage to human bronchial epithelial cells, Toxicology, vol.213, pp.66-73, 2005.

S. Halappanavar, P. Jackson, A. Williams, K. A. Jensen, K. S. Hougaard et al., Pulmonary response to surface-coated nanotitanium dioxide particles includes induction of acute phase response genes, inflammatory cascades, and changes in microRNAs: a toxicogenomic study, Environmental and molecular mutagenesis, vol.52, pp.425-439, 2011.

S. Halappanavar, A. T. Saber, N. Decan, K. A. Jensen, D. Wu et al., Transcriptional profiling identifies physicochemical properties of nanomaterials that are determinants of the in vivo pulmonary response, Environmental and molecular mutagenesis, vol.56, pp.245-264, 2015.

H. K. Hamadeh, R. P. Amin, R. S. Paules, and C. A. Afshari, An overview of toxicogenomics, Current issues in molecular biology, vol.4, pp.45-56, 2002.

K. Hamai, H. Iwamoto, N. Ishikawa, Y. Horimasu, T. Masuda et al., Comparative Study of Circulating MMP-7, CCL18, KL-6, SP-A, and SP-D as Disease Markers of Idiopathic Pulmonary Fibrosis, p.4759040, 2016.

N. Hambly, C. Shimbori, and M. Kolb, Molecular classification of idiopathic pulmonary fibrosis: personalized medicine, genetics and biomarkers, Respirology, vol.20, pp.1010-1022, 2015.

R. F. Hamilton, N. Wu, D. Porter, M. Buford, W. M. Holian et al., Particle length-dependent titanium dioxide nanomaterials toxicity and bioactivity, Particle and fibre toxicology, vol.6, p.35, 2009.

W. H. Heijne, A. S. Kienhuis, B. Van-ommen, S. Rh, and J. P. Groten, Systems toxicology: applications of toxicogenomics, transcriptomics, proteomics and metabolomics in toxicology, Expert review of proteomics, vol.2, pp.767-780, 2005.

U. Heinrich, R. Fuhst, S. Rittinghausen, O. Creutzenberg, B. Bellmann et al., Chronic Inhalation Exposure of Wistar Rats and 2 Different Strains of Mice to Diesel-Engine Exhaust, Carbon-Black, and Titanium-Dioxide, Inhalation toxicology, vol.7, pp.533-556, 1995.

N. Herlin-boime, I. Michaud-soret, C. Fauquant, A. L. Carrière, and M. , De la synthèse des nanoparticules de TiO2 à l'étude de leur comportement, Biofutur, vol.294, p.3506, 2013.

S. Herold, M. K. Lohmeyer, and J. , Acute lung injury: how macrophages orchestrate resolution of inflammation and tissue repair, Frontiers in immunology, vol.2, p.65, 2011.

B. Hervé-bazin, Les nanoparticules: Un enjeu majeur pour la santé au travail ?, 2007.

P. M. Hext, J. A. Tomenson, and P. Thompson, Titanium dioxide: inhalation toxicology and epidemiology, The Annals of occupational hygiene, vol.49, pp.461-472, 2005.

J. G. Hirsch, Bactericidal action of histone, The Journal of experimental medicine, vol.108, pp.925-944, 1958.

S. M. Ho, A. Johnson, P. Tarapore, V. Janakiram, X. Zhang et al., Environmental epigenetics and its implication on disease risk and health outcomes, ILAR journal, vol.53, pp.289-305, 2012.

D. Hohr, Y. Steinfartz, R. P. Schins, A. M. Knaapen, G. Martra et al., The surface area rather than the surface coating determines the acute inflammatory response after instillation of fine and ultrafine TiO2 in the rat, International journal of hygiene and environmental health, vol.205, pp.239-244, 2002.

K. S. Hougaard, P. Jackson, K. A. Jensen, J. J. Sloth, K. Loschner et al., Effects of prenatal exposure to surface-coated nanosized titanium dioxide (UV-Titan). A study in mice, Particle and fibre toxicology, vol.7, p.16, 2010.

H. Il and Y. J. Huang, Effects of various physicochemical characteristics on the toxicities of ZnO and TiO nanoparticles toward human lung epithelial cells, The Science of the total environment, vol.409, pp.1219-1228, 2011.

R. Hu, X. Gong, Y. Duan, N. Li, Y. Che et al., Neurotoxicological effects and the impairment of spatial recognition memory in mice caused by exposure to TiO2 nanoparticles, Biomaterials, vol.31, pp.8043-8050, 2010.

C. Huang, M. Sun, Y. Yang, F. Wang, X. Ma et al., Titanium dioxide nanoparticles prime a specific activation state of macrophages, Nanotoxicology, vol.11, pp.737-750, 2017.

D. W. Huang, S. Bt, and R. A. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nature protocols, vol.4, pp.44-57, 2009.

M. Husain, Z. O. Kyjovska, J. Bourdon-lacombe, A. T. Saber, K. A. Jensen et al., Carbon black nanoparticles induce biphasic gene expression changes associated with inflammatory responses in the lungs of C57BL/6 mice following a single intratracheal instillation, Toxicology and applied pharmacology, vol.289, pp.573-588, 2015.

M. Husain, A. T. Saber, C. Guo, N. R. Jacobsen, K. A. Jensen et al.,

, Pulmonary instillation of low doses of titanium dioxide nanoparticles in mice leads to particle retention and gene expression changes in the absence of inflammation, Toxicology and applied pharmacology, vol.269, pp.250-262

. Iarc, Carbon black, titanium dioxide, and talc, 2010.

. Iarc, Carbon Black, Titanium Dioxide, and Talc., 93. International Agency for Research on Cancer Monographs on the Evaluation of Carcinogenic Risks to Human, 2010.

S. Ichihara, W. Li, S. Omura, Y. Fujitani, Y. Liu et al., Effects on respiratory and cardiovascular systems in workers handling titanium dioxide particles, The 6th international symposium on nanotechnology, 2013.

I. Icrp, ICRP publication 66: human respiratory tract model for radiological protection, 1994.

R. Ilsi, The relevance of the rat lung response to particle overload for human risk assessment: a workshop consensus report, Inhalation toxicology, vol.12, p.1, 2000.

. Inra, Additif alimentaire E171 : les premiers résultats de l'exposition orale aux nanoparticules de dioxyde de titane, Journal, 2017.

. Inrs, Utilisation du dioxyde de titane nanométrique. Cas particulier de la filière BTP, 2012.

. Inrs, Dioxyde de titane, Fiche toxicologique n°291, Journal, 2013.

, ISO/TR 13014:2012, Nanotechnologies -Guidance on physicochemical characterization of engineered nanoscale materials for toxicologic assessment, 2012.

, Nanotechnology -Nanoparticles in powder form -Characteristics and measurements, 2013.

, ISO/TS 80004-2: 2015 Nanotechnologies-Vocabulary-Part, vol.2, 2015.

, ISO/TC 229 -Nanotechnologies, 2016.

R. Jansen, U. Osterwalder, S. Q. Wang, M. Burnett, and H. W. Lim, Photoprotection: part II. Sunscreen: development, efficacy, and controversies, Journal of the American Academy of Dermatology, vol.69, pp.881-862, 2013.

J. H. Ji, J. H. Jung, S. S. Kim, J. U. Yoon, J. D. Park et al., Twenty-eight-day inhalation toxicity study of silver nanoparticles in Sprague-Dawley rats, Inhalation toxicology, vol.19, pp.857-871, 2007.

K. Jones, J. Morton, I. Smith, K. Jurkschat, A. H. Harding et al., Human in vivo and in vitro studies on gastrointestinal absorption of titanium dioxide nanoparticles, Toxicology letters, vol.233, pp.95-101, 2015.

B. Jovanovic, L. Anastasova, E. W. Rowe, Y. Zhang, C. Ar et al., Effects of nanosized titanium dioxide on innate immune system of fathead minnow (Pimephales promelas Rafinesque, 1820), Ecotoxicology and environmental safety, vol.74, pp.675-683, 2011.

H. Kan, Z. Wu, Y. C. Lin, T. H. Chen, J. L. Cumpston et al., The role of nodose ganglia in the regulation of cardiovascular function following pulmonary exposure to ultrafine titanium dioxide, Nanotoxicology, vol.8, pp.447-454, 2014.

S. J. Kang, B. M. Kim, Y. J. Lee, and C. Hw, Titanium dioxide nanoparticles trigger p53-mediated damage response in peripheral blood lymphocytes, Environmental and molecular mutagenesis, vol.49, pp.399-405, 2008.

K. Kettler, K. Veltman, D. Van-de-meent, V. Wezel, and A. J. Hendriks, Cellular uptake of nanoparticles as determined by particle properties, experimental conditions, and cell type, Environmental toxicology and chemistry, vol.33, pp.481-492, 2014.

N. Khansari, Y. Shakiba, and M. Mahmoudi, Chronic inflammation and oxidative stress as a major cause of agerelated diseases and cancer, Recent patents on inflammation & allergy drug discovery, vol.3, pp.73-80, 2009.

J. K. Kim, S. M. Pradhan, and S. , Epigenetic mechanisms in mammals. Cellular and molecular life sciences, CMLS, vol.66, pp.596-612, 2009.

S. C. Kim, D. R. Chen, C. Qi, R. M. Gelein, J. N. Finkelstein et al., A nanoparticle dispersion method for in vitro and in vivo nanotoxicity study, Nanotoxicology, vol.4, pp.42-51, 2010.

T. L. Knuckles, J. Yi, D. G. Frazer, H. D. Leonard, B. T. Chen et al., Nanoparticle inhalation alters systemic arteriolar vasoreactivity through sympathetic and cyclooxygenase-mediated pathways, Nanotoxicology, vol.6, pp.724-735, 2012.

A. Koizumi, M. Tsukada, S. Hirano, S. Kamiyama, H. Masuda et al., Energy Restriction That Inhibits Cellular Proliferation by Torpor Can Decrease Susceptibility to Spontaneous and Asbestos-Induced Lung-Tumors in a, /J Mice. Lab Invest, vol.68, pp.728-739, 1993.

W. G. Kreyling, Discovery of unique and ENM-specific pathophysiologic pathways: Comparison of the translocation of inhaled iridium nanoparticles from nasal epithelium versus alveolar epithelium towards the brain of rats, Toxicology and applied pharmacology, vol.299, pp.41-46, 2016.

W. G. Kreyling, M. Semmler-behnke, J. Seitz, W. Scymczak, A. Wenk et al., Size dependence of the translocation of inhaled iridium and carbon nanoparticle aggregates from the lung of rats to the blood and secondary target organs, Inhalation toxicology, vol.21, pp.55-60, 2009.

W. G. Kreyling, M. Semmler-behnke, T. S. Moller, and W. , Differences in the biokinetics of inhaled nanoversus micrometer-sized particles, Accounts of chemical research, vol.46, pp.714-722, 2013.

W. G. Kreyling, M. Semmler, F. Erbe, P. Mayer, S. Takenaka et al., Translocation of ultrafine insoluble iridium particles from lung epithelium to extrapulmonary organs is size dependent but very low, Journal of toxicology and environmental health Part A, vol.65, pp.1513-1530, 2002.

P. Kulkarni, P. A. Baron, and K. Willeke, Aerosol Measurement: Principles, Techniques, and Applications, 2011.

V. Kumar and A. Sharma, Neutrophils: Cinderella of innate immune system, International immunopharmacology, vol.10, pp.1325-1334, 2010.

J. Labille, J. Feng, C. Botta, D. Borschneck, M. Sammut et al., Aging of TiO(2) nanocomposites used in sunscreen. Dispersion and fate of the degradation products in aqueous environment, Environ Pollut, vol.158, pp.3482-3489, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01519536

S. Lacour, Définir les nanomatériaux : une controverse scientifique ou normative ? ANSES -Bulletin de veille scientifique -Santé Environnement Travail 16, 2011.

R. Landsiedel, E. Fabian, L. Ma-hock, W. Wohlleben, K. Wiench et al., Toxico-/biokinetics of nanomaterials, Archives of toxicology, vol.86, pp.1021-1060, 2012.

F. Larese-filon, M. Mauro, G. Adami, B. M. Crosera, and M. , Nanoparticles skin absorption: New aspects for a safety profile evaluation. Regulatory toxicology and pharmacology, RTP, vol.72, pp.310-322, 2015.

A. J. Leblanc, A. M. Moseley, B. T. Chen, D. Frazer, C. V. Nurkiewicz et al., Nanoparticle inhalation impairs coronary microvascular reactivity via a local reactive oxygen species-dependent mechanism, Cardiovascular toxicology, vol.10, pp.27-36, 2010.

C. G. Lee, Chitin, chitinases and chitinase-like proteins in allergic inflammation and tissue remodeling, Yonsei medical journal, vol.50, pp.22-30, 2009.

K. P. Lee, T. Hj, and C. F. Reinhardt, Pulmonary response of rats exposed to titanium dioxide (TiO2) by inhalation for two years, Toxicology and applied pharmacology, vol.79, pp.179-192, 1985.

G. Li, L. Lv, H. Fan, J. Ma, Y. Li et al., Effect of the agglomeration of TiO2 nanoparticles on their photocatalytic performance in the aqueous phase, Journal of colloid and interface science, vol.348, pp.342-347, 2010.

N. Li, S. Georas, A. N. Fritz, P. Xia, T. Williams et al., A work group report on ultrafine particles (American Academy of Allergy, Asthma & Immunology): Why ambient ultrafine and engineered nanoparticles should receive special attention for possible adverse health outcomes in human subjects, Journal of Allergy and Clinical Immunology, vol.138, pp.386-396, 2016.

Y. Li, J. Li, J. Yin, W. Li, C. Kang et al., Systematic influence induced by 3 nm titanium dioxide following intratracheal instillation of mice, Journal of nanoscience and nanotechnology, vol.10, pp.8544-8549, 2010.

Y. Li, J. Yan, W. Ding, Y. Chen, P. Lm et al., Genotoxicity and gene expression analyses of liver and lung tissues of mice treated with titanium dioxide nanoparticles, Mutagenesis, vol.32, pp.33-46, 2017.

M. Liao and H. Liu, Gene expression profiling of nephrotoxicity from copper nanoparticles in rats after repeated oral administration, Environmental toxicology and pharmacology, vol.34, pp.67-80, 2012.

S. H. Liou, C. S. Tsai, D. Pelclova, M. K. Schubauer-berigan, and P. A. Schulte, Assessing the first wave of epidemiological studies of nanomaterial workers, vol.17, p.413, 2015.

R. Liu, L. Yin, Y. Pu, G. Liang, J. Zhang et al., Pulmonary toxicity induced by three forms of titanium dioxide nanoparticles via intra-tracheal instillation in rats, Progress in Natural Science, vol.19, pp.573-579, 2009.

R. Liz, J. C. Simard, L. Lb, and D. Girard, Silver nanoparticles rapidly induce atypical human neutrophil cell death by a process involving inflammatory caspases and reactive oxygen species and induce neutrophil extracellular traps release upon cell adhesion, International immunopharmacology, vol.28, pp.616-625, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01352190

L. Ma-hock, S. Burkhardt, V. Strauss, A. O. Gamer, K. Wiench et al., Development of a short-term inhalation test in the rat using nano-titanium dioxide as a model substance, Inhalation toxicology, vol.21, pp.102-118, 2009.

L. Ma-hock, A. O. Gamer, R. Landsiedel, E. Leibold, T. Frechen et al., Generation and characterization of test atmospheres with nanomaterials, Inhalation toxicology, vol.19, pp.833-848, 2007.

Z. Magdolenova, A. Collins, A. Kumar, A. Dhawan, V. Stone et al., Mechanisms of genotoxicity. A review of in vitro and in vivo studies with engineered nanoparticles, Nanotoxicology, vol.8, pp.233-278, 2014.

M. Fo and G. S. , The M1 and M2 paradigm of macrophage activation: time for reassessment, F1000prime reports, vol.6, p.13, 2014.

T. N. Mayadas, C. X. Lowell, and C. A. , The multifaceted functions of neutrophils, Annual review of pathology, vol.9, pp.181-218, 2014.

H. Meng, T. Xia, G. S. Nel, and A. E. , A Predictive Toxicological Paradigm for the Safety Assessment of Nanomaterials, ACS nano, vol.3, pp.1620-1627, 2009.

F. J. Miller, Dosimetry of particles in laboratory animals and humans in relationship to issues surrounding lung overload and human health risk assessment: a critical review, Inhalation toxicology, vol.12, pp.19-57, 2000.

C. D. Mills, K. Kincaid, J. M. Alt, H. Mj, and A. M. Hill, M-1/M-2 macrophages and the Th1/Th2 paradigm, J Immunol, vol.164, pp.6166-6173, 2000.
DOI : 10.4049/jimmunol.164.12.6166

URL : http://www.jimmunol.org/content/164/12/6166.full.pdf

B. B. Misra, New tools and resources in metabolomics, pp.2016-2017, 2018.
DOI : 10.1002/elps.201700441

P. Moller, D. M. Jensen, R. S. Wils, M. Andersen, D. Ph et al., Assessment of evidence for nanosized titanium dioxide-generated DNA strand breaks and oxidatively damaged DNA in cells and animal models, Nanotoxicology, vol.11, pp.1237-1256, 2017.

C. Monteiller, L. Tran, W. Macnee, S. Faux, A. Jones et al., The pro-inflammatory effects of low-toxicity low-solubility particles, nanoparticles and fine particles, on epithelial cells in vitro: the role of surface area, Occupational and environmental medicine, vol.64, pp.609-615, 2007.

Y. Morimoto, M. Horie, N. Kobayashi, S. N. Shimada, and M. , Inhalation toxicity assessment of carbon-based nanoparticles, Accounts of chemical research, vol.46, pp.770-781, 2013.

P. E. Morrow, Possible mechanisms to explain dust overloading of the lungs, Fundamental and applied toxicology : official journal of the Society of Toxicology, vol.10, pp.369-384, 1988.

A. R. Moschen, T. E. Adolph, R. R. Gerner, W. V. Tilg, and H. , Lipocalin-2: A Master Mediator of Intestinal and Metabolic Inflammation, Trends in endocrinology and metabolism: TEM, vol.28, pp.388-397, 2017.

P. J. Murray, J. E. Allen, S. K. Biswas, E. A. Fisher, D. W. Gilroy et al., Macrophage activation and polarization: nomenclature and experimental guidelines, Immunity, vol.41, pp.14-20, 2014.
DOI : 10.1016/j.immuni.2014.07.009

URL : https://doi.org/10.1016/j.immuni.2014.07.009

M. Pj and T. A. Wynn, Protective and pathogenic functions of macrophage subsets, Nature reviews Immunology, vol.11, pp.723-737, 2011.

. Nanoderm, Quality of skin as a barrier to ultra-fine particles (Final report), 2007.

S. P. Narciso, E. Nadziejko, L. C. Chen, G. T. Nadziejko, and C. , Adaptation to stress induced by restraining rats and mice in nose-only inhalation holders, Inhalation toxicology, vol.15, pp.1133-1143, 2003.
DOI : 10.1080/713857296

M. Naya, N. Kobayashi, M. Ema, S. Kasamoto, M. Fukumuro et al., vivo genotoxicity study of titanium dioxide nanoparticles using comet assay following intratracheal instillation in rats. Regulatory toxicology and pharmacology, vol.62, pp.1-6, 2012.

A. Nel, T. Xia, M. L. , and L. N. , Toxic Potential of Materials at the Nanolevel, Science, vol.311, pp.622-627, 2006.

M. D. Newman, S. M. Ellis, and J. I. , The safety of nanosized particles in titanium dioxide-and zinc oxide-based sunscreens, Journal of the American Academy of Dermatology, vol.61, pp.685-692, 2009.

J. K. Nicholson, J. C. Lindon, and E. Holmes, Metabonomics': understanding the metabolic responses of living systems to pathophysiological stimuli via multivariate statistical analysis of biological NMR spectroscopic data. Xenobiotica; the fate of foreign compounds in biological systems, vol.29, pp.1181-1189, 1999.

J. Nikota, A. Williams, C. L. Yauk, H. Wallin, U. Vogel et al., Meta-analysis of transcriptomic responses as a means to identify pulmonary disease outcomes for engineered nanomaterials, Particle and fibre toxicology, vol.13, p.25, 2016.

. Niosh, Occupational Exposure to Titanium Dioxide Current Intelligence. National Institute for Occupational Safety and Health Bulletin 63, 2011.

A. Noel, M. Charbonneau, Y. Cloutier, R. Tardif, and G. Truchon, Rat pulmonary responses to inhaled nano-TiO(2): effect of primary particle size and agglomeration state, Particle and fibre toxicology, vol.10, p.48, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-01132013

A. Noel, Y. Cloutier, K. J. Wilkinson, C. Dion, S. Halle et al., Generating nanoaerosols from TiO(2) (5 nm) nanoparticles showing different agglomeration states. Application to toxicological studies, Journal of occupational and environmental hygiene, vol.10, pp.86-96, 2013.

T. R. Nurkiewicz, D. W. Porter, A. F. Hubbs, J. L. Cumpston, B. T. Chen et al., Nanoparticle inhalation augments particle-dependent systemic microvascular dysfunction, Toxicological sciences : an official journal of the Society of Toxicology, vol.5, issue.1, pp.191-203, 2008.

T. R. Nurkiewicz, D. W. Porter, A. F. Hubbs, S. Stone, A. M. Moseley et al., Pulmonary particulate matter and systemic microvascular dysfunction, Res Rep Health Eff Inst, pp.3-48, 2011.

E. F. Nuwaysir, M. Bittner, J. Trent, J. C. Barrett, and C. A. Afshari, Microarrays and toxicology: the advent of toxicogenomics, Molecular carcinogenesis, vol.24, pp.153-159, 1999.

M. O'brien, The reciprocal relationship between inflammation and coagulation, Topics in companion animal medicine, vol.27, pp.46-52, 2012.

S. O'reilly, Epigenetics in fibrosis. Molecular aspects of medicine, vol.54, pp.89-102, 2017.

G. Oberdorster, Lung particle overload: implications for occupational exposures to particles. Regulatory toxicology and pharmacology, RTP, vol.21, pp.123-135, 1995.

G. Oberdorster, Significance of particle parameters in the evaluation of exposure-dose-response relationships of inhaled particles, Inhalation toxicology, vol.8, pp.73-89, 1996.

G. Oberdorster, Pulmonary effects of inhaled ultrafine particles, International archives of occupational and environmental health, vol.74, pp.1-8, 2001.

G. Oberdorster, J. Ferin, R. Gelein, S. Sc, and J. Finkelstein, Role of the alveolar macrophage in lung injury: studies with ultrafine particles, Environmental health perspectives, vol.97, pp.193-199, 1992.

G. Oberdorster, F. J. Lehnert, and B. E. , Correlation between particle size, in vivo particle persistence, and lung injury, Environmental health perspectives, vol.102, pp.173-179, 1994.

G. Oberdorster, O. E. Oberdorster, and J. , Nanotoxicology: an emerging discipline evolving from studies of ultrafine particles, Environmental health perspectives, vol.113, pp.823-839, 2005.

. Oecd, Dossier on Titanium Dioxide -Part, vol.1, pp.3-105, 2015.

. Oecd, Physical-chemical parameters : measurements and methods relevant fo the regulation of nanomaterials -OECD Workshop Report -Series on the Safety of Manufactured Nanomaterials No. 63, Publications in the Series on the Safety of Manufactured Nanomaterials, 2016.

. Oecd, Organization for Economic Co-operation and Development Guidelines for the Testing of Chemicals, 2017.

P. Oh, P. Borgstrom, H. Witkiewicz, Y. Li, B. J. Borgstrom et al., Live dynamic imaging of caveolae pumping targeted antibody rapidly and specifically across endothelium in the lung, Nature biotechnology, vol.25, pp.327-337, 2007.

J. Okuda-shimazaki, S. Takaku, K. Kanehira, S. S. Taniguchi, and A. , Effects of titanium dioxide nanoparticle aggregate size on gene expression, International journal of molecular sciences, vol.11, pp.2383-2392, 2010.

M. F. Oleksiak, Toxicogenomics, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02095321

O. S. , The relevance of the rat lung response to particle overload for human risk assessment: a workshop consensus report, Inhalation toxicology. ILSI Risk Science Institute, pp.1-17, 2000.

M. L. Ostraat, K. A. Swain, and J. J. Krajewski, SiO2 aerosol nanoparticle reactor for occupational health and safety studies, Journal of occupational and environmental hygiene, vol.5, pp.390-398, 2008.

T. Oyabu, Y. Morimoto, H. Izumi, Y. Yoshiura, T. Tomonaga et al., Comparison between whole-body inhalation and nose-only inhalation on the deposition and health effects of nanoparticles, vol.21, pp.42-48, 2016.

N. M. Page, Hemokinins and endokinins. Cellular and molecular life sciences, CMLS, vol.61, pp.1652-1663, 2004.

V. Papayannopoulos, K. D. Metzler, H. A. Zychlinsky, and A. , Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps, The Journal of cell biology, vol.191, pp.677-691, 2010.

A. Pardo, S. Cabrera, M. Maldonado, and M. Selman, Role of matrix metalloproteinases in the pathogenesis of idiopathic pulmonary fibrosis, Respiratory research, vol.17, p.23, 2016.

R. Pati, I. Das, R. K. Mehta, S. R. Sonawane, and A. , Zinc-Oxide Nanoparticles Exhibit Genotoxic, Clastogenic, Cytotoxic and Actin Depolymerization Effects by Inducing Oxidative Stress Responses in Macrophages and Adult Mice, Toxicological sciences : an official journal of the Society of Toxicology, vol.150, pp.454-472, 2016.

D. Pelclova, V. Zdimal, Z. Fenclova, S. Vlckova, J. Schwarz et al.,

, Markers of oxidative stress are elevated in workers exposed to nanoparticles, Proceedings of the 4th International Conference on NANOCON, pp.23-25

D. Pelclova, V. Zdimal, Z. Fenclova, S. Vlckova, F. Turci et al., Markers of oxidative damage of nucleic acids and proteins among workers exposed to TiO2 (nano) particles, Occupational and environmental medicine, vol.73, pp.110-118, 2016.

D. Pelclova, V. Zdimal, P. Kacer, S. Vlckova, Z. Fenclova et al., Markers of nucleic acids and proteins oxidation among office workers exposed to air pollutants including (nano)TiO2 particles, Neuro endocrinology letters, vol.37, pp.13-16, 2016.

D. Pelclova, V. Zdimal, P. Kacer, N. Zikova, M. Komarc et al., Markers of lipid oxidative damage in the exhaled breath condensate of nano TiO2 production workers, Nanotoxicology, vol.11, pp.52-63, 2017.

J. N. Pennefather, A. Lecci, M. L. Candenas, E. Patak, F. M. Pinto et al., Tachykinins and tachykinin receptors: a growing family, Life sciences, vol.74, pp.1445-1463, 2004.

J. Petaja, Inflammation and coagulation. An overview, Thrombosis research, vol.127, pp.34-37, 2011.

C. Pineau, La protéomique : nouvelles approches en génomique fonctionnelle. MT médecine de la reproduction, vol.8, pp.373-384, 2006.

C. Pisani, J. C. Gaillard, V. Nouvel, M. Odorico, A. et al., High-throughput, quantitative assessment of the effects of low-dose silica nanoparticles on lung cells: grasping complex toxicity with a great depth of field, BMC genomics, vol.16, p.315, 2015.

D. W. Porter, N. Wu, A. F. Hubbs, R. R. Mercer, K. Funk et al., Differential mouse pulmonary dose and time course responses to titanium dioxide nanospheres and nanobelts, Toxicological sciences : an official journal of the Society of Toxicology, vol.131, pp.179-193, 2013.

P. Bn and R. T. Stein, Neutrophil Extracellular Traps in Pulmonary Diseases: Too Much of a Good Thing, Frontiers in immunology, vol.7, p.311, 2016.

P. Roller and M. , Carcinogenicity study with nineteen granular dusts in rats, Eur J Oncol, vol.10, pp.249-281, 2005.

L. R. Potter, A. -. Dickey, and D. M. , Natriuretic peptides, their receptors, and cyclic guanosine monophosphate-dependent signaling functions, Endocrine reviews, vol.27, pp.47-72, 2005.

S. S. Poulsen, N. R. Jacobsen, S. Labib, D. Wu, M. Husain et al.,

, Transcriptomic analysis reveals novel mechanistic insight into murine biological responses to multi-walled carbon nanotubes in lungs and cultured lung epithelial cells, PloS one, vol.8, p.80452

S. S. Poulsen, A. T. Saber, A. Mortensen, J. Szarek, D. Wu et al., Changes in cholesterol homeostasis and acute phase response link pulmonary exposure to multi-walled carbon nanotubes to risk of cardiovascular disease, Toxicology and applied pharmacology, vol.283, pp.210-222, 2015.

S. S. Poulsen, A. T. Saber, A. Williams, O. Andersen, C. Kobler et al., MWCNTs of different physicochemical properties cause similar inflammatory responses, but differences in transcriptional and histological markers of fibrosis in mouse lungs, Toxicology and applied pharmacology, vol.284, pp.16-32, 2015.

O. Price, B. Asgharian, F. Miller, C. F. , D. Winter-sorkina et al., Multiple Path Particle Dosimetry model (MPPD v1. 0): A model for human and rat airway particle dosimetry, 2002.

R. , Éléments issus des déclarations des substances à l'état nanoparticulaire: rapport d'étude 2017. Ministère de la Transition écologique et solidaire, p.317, 2017.

L. Rahman, D. Wu, M. Johnston, W. A. Halappanavar, and S. , Toxicogenomics analysis of mouse lung responses following exposure to titanium dioxide nanomaterials reveal their disease potential at high doses, Mutagenesis, vol.32, pp.59-76, 2017.

A. Ramesh, S. S. Varghese, D. J. Malaiappan, and S. , Role of sulfiredoxin in systemic diseases influenced by oxidative stress, Redox biology, vol.2, pp.1023-1028, 2014.

C. Relier, M. Dubreuil, L. Garcia, O. Cordelli, E. Mejia et al., Study of TiO2 P25 Nanoparticles Genotoxicity on Lung, Blood, and Liver Cells in Lung Overload and Non-Overload Conditions After Repeated Respiratory Exposure in Rats, Toxicological sciences : an official journal of the Society of Toxicology, vol.156, pp.527-537, 2017.
URL : https://hal.archives-ouvertes.fr/ineris-01853471

W. Ren, M. Z. Iqbal, L. Zeng, T. Chen, Y. Pan et al., Black TiO2 based core-shell nanocomposites as doxorubicin carriers for thermal imaging guided synergistic therapy of breast cancer, Nanoscale, vol.9, pp.11195-11204, 2017.

R. Witschger and O. , Les nanomatériaux. Définitions, risques toxicologiques, caractérisation de l'exposition professionnelle et mesures de prévention, ED, vol.6050, p.48, 2012.

A. J. Rickard and M. J. Young, Corticosteroid receptors, macrophages and cardiovascular disease, Journal of molecular endocrinology, vol.42, pp.449-459, 2009.

B. Rihn, C. Saliou, M. C. Bottin, K. G. Packer, and L. , From ancient remedies to modern therapeutics: pine bark uses in skin disorders revisited, Phytotherapy Research: An International Journal Devoted to Pharmacological and Toxicological Evaluation of Natural Product Derivatives, vol.15, pp.76-78, 2001.

K. R. Rogers, C. J. Morris, and D. R. Blake, The cytoskeleton and its importance as a mediator of inflammation, Annals of the rheumatic diseases, vol.51, pp.565-571, 1992.

M. Roursgaard, K. A. Jensen, S. S. Poulsen, N. E. Jensen, L. K. Poulsen et al., Acute and subchronic airway inflammation after intratracheal instillation of quartz and titanium dioxide agglomerates in mice, TheScientificWorldJournal, vol.11, pp.801-825, 2011.

R. Wms and R. L. Burch, The principles of humane experimental technique, Journal, 1959.

N. Sadrieh, A. M. Wokovich, N. V. Gopee, J. Zheng, D. Haines et al., Lack of significant dermal penetration of titanium dioxide from sunscreen formulations containing nano-and submicron-size TiO2 particles, Toxicological sciences : an official journal of the Society of Toxicology, vol.115, pp.156-166, 2010.

Y. Sagawa, M. Futakuchi, J. Xu, K. Fukamachi, Y. Sakai et al., Lack of promoting effect of titanium dioxide particles on chemically-induced skin carcinogenesis in rats and mice, The Journal of toxicological sciences, vol.37, pp.317-327, 2012.

. Sager and V. Castranova, Surface area of particle administered versus mass in determining the pulmonary toxicity of ultrafine and fine carbon black: comparison to ultrafine titanium dioxide, Particle and fibre toxicology, vol.6, p.15, 2009.

T. M. Sager, K. C. Castranova, and V. , Pulmonary response to intratracheal instillation of ultrafine versus fine titanium dioxide: role of particle surface area, Particle and fibre toxicology, vol.5, p.17, 2008.

X. Sang, L. Zheng, Q. Sun, N. Li, Y. Cui et al., The chronic spleen injury of mice following long-term exposure to titanium dioxide nanoparticles, Journal of biomedical materials research Part A, vol.100, pp.894-902, 2012.

C. M. Sayes, R. Wahi, P. A. Kurian, Y. Liu, J. L. West et al., Correlating nanoscale titania structure with toxicity: a cytotoxicity and inflammatory response study with human dermal fibroblasts and human lung epithelial cells, Toxicological sciences : an official journal of the Society of Toxicology, vol.92, pp.174-185, 2006.

I. J. Scherer, H. Pv, and R. B. Harris, The importance of corticosterone in mediating restraint-induced weight loss in rats, Physiology & behavior, vol.102, pp.225-233, 2011.

. Schins and A. M. Knaapen, Genotoxicity of poorly soluble particles, Inhalation toxicology, vol.19, pp.189-198, 2007.

A. H. Schmaier, The contact activation and kallikrein/kinin systems: pathophysiologic and physiologic activities, Journal of thrombosis and haemostasis : JTH, vol.14, pp.28-39, 2016.

M. Senzui, T. Tamura, K. Miura, Y. Ikarashi, Y. Watanabe et al., Study on penetration of titanium dioxide (TiO(2)) nanoparticles into intact and damaged skin in vitro, The Journal of toxicological sciences, vol.35, pp.107-113, 2010.

N. B. Shah, G. M. Vercellotti, J. G. White, A. Fegan, . Wagner-cr et al., Blood-nanoparticle interactions and in vivo biodistribution: impact of surface PEG and ligand properties, Molecular pharmaceutics, vol.9, pp.2146-2155, 2012.

P. W. Shaul, Regulation of endothelial nitric oxide synthase: location, location, location, Annual review of physiology, vol.64, pp.749-774, 2002.

H. Shi, R. Magaye, C. V. Zhao, and J. , Titanium dioxide nanoparticles: a review of current toxicological data, Particle and fibre toxicology, vol.10, p.15, 2013.

R. K. Shukla, V. Sharma, A. K. Pandey, S. Singh, S. Sultana et al., ROS-mediated genotoxicity induced by titanium dioxide nanoparticles in human epidermal cells, Toxicology in vitro : an international journal published in association with BIBRA, vol.25, pp.231-241, 2011.

S. Shyamasundar, C. T. Ng, L. Y. Yung, D. St, and B. H. Bay, Epigenetic mechanisms in nanomaterial-induced toxicity, Epigenomics, vol.7, pp.395-411, 2015.

J. Siemiatycki, L. Richardson, L. Nadon, R. Lakhani, D. Begin et al., Risk factors for cancer in the workplace, 1991.

J. R. Smith, Bioinformatics and the eye, Journal of ocular biology, vol.2, pp.161-163, 2010.

B. Smolkova, N. El-yamani, A. R. Collins, G. Ac, and M. Dusinska, Epigenetic changes induced by nanomaterials and possible impact on health. Food and chemical toxicology : an international journal published for the, British Industrial Biological Research Association, vol.77, pp.64-73, 2015.

M. B. Snipes, O. Tr, and H. C. Yeh, Deposition and retention patterns for 3-, 9-, and 15-micron latex microspheres inhaled by rats and guinea pigs, Experimental lung research, vol.14, pp.37-50, 1988.

B. N. Snyder-talkington, J. Dymacek, D. W. Porter, M. G. Wolfarth, R. R. Mercer et al., System-based identification of toxicity pathways associated with multi-walled carbon nanotube-induced pathological responses, Toxicology and applied pharmacology, vol.272, pp.476-489, 2013.

B. Song, T. Zhou, W. Yang, J. Liu, and L. Shao, Contribution of oxidative stress to TiO2 nanoparticle-induced toxicity, Environmental toxicology and pharmacology, vol.48, pp.130-140, 2016.

P. A. Stapleton, Q. A. Hathaway, C. E. Nichols, A. B. Abukabda, M. V. Pinti et al., Maternal engineered nanomaterial inhalation during gestation alters the fetal transcriptome, Particle and fibre toxicology, vol.15, p.3, 2018.

F. Stenback, R. J. Sellakumar, and A. , Carcinogenicity of Benzo(a)pyrene and dusts in the hamster lung (instilled intratracheally with titanium oxide, aluminum oxide, carbon and ferric oxide), Oncology, vol.33, pp.29-34, 1976.

A. Stoccoro, H. L. Karlsson, C. F. Migliore, and L. , Epigenetic effects of nano-sized materials, Toxicology, vol.313, pp.3-14, 2013.

S. J. Sturla, A. R. Boobis, R. E. Fitzgerald, J. Hoeng, R. J. Kavlock et al., Systems toxicology: from basic research to risk assessment, Chemical research in toxicology, vol.27, pp.314-329, 2014.

L. P. Sycheva, V. S. Zhurkov, V. V. Iurchenko, N. O. Daugel-dauge, M. A. Kovalenko et al., Investigation of genotoxic and cytotoxic effects of micro-and nanosized titanium dioxide in six organs of mice in vivo, Mutation research, vol.726, pp.8-14, 2011.

D. Szklarczyk, A. Franceschini, S. Wyder, K. Forslund, D. Heller et al., STRING v10: protein-protein interaction networks, integrated over the tree of life, Nucleic acids research, vol.43, pp.447-452, 2015.

D. Szklarczyk, J. H. Morris, H. Cook, M. Kuhn, S. Wyder et al., The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible, Nucleic acids research, vol.45, pp.362-368, 2017.

K. Takeda, K. I. Suzuki, A. Ishihara, M. Kubo-irie, R. Fujimoto et al., Nanoparticles Transferred from Pregnant Mice to Their Offspring Can Damage the Genital and Cranial Nerve Systems, J Health Sci, vol.55, pp.95-102, 2009.

M. H. Tan, C. A. Commens, L. Burnett, and P. J. Snitch, A pilot study on the percutaneous absorption of microfine titanium dioxide from sunscreens, The Australasian journal of dermatology, vol.37, pp.185-187, 1996.

C. F. Taylor, N. W. Paton, K. S. Lilley, P. A. Binz, R. K. Julian et al., The minimum information about a proteomics experiment (MIAPE), Nature biotechnology, vol.25, pp.887-893, 2007.

. Tga, Literature Review on the safety of titanium dioxide and zinc oxide nanoparticles in sunscreens, 2016.

S. C. Tilton, N. J. Karin, A. Tolic, Y. Xie, X. Lai et al., Three human cell types respond to multi-walled carbon nanotubes and titanium dioxide nanobelts with cell-specific transcriptomic and proteomic expression patterns, Nanotoxicology, vol.8, pp.533-548, 2014.

A. A. Torrano, Â. S. Pereira, O. N. Oliveira, and A. Barros-timmons, Probing the interaction of oppositely charged gold nanoparticles with DPPG and DPPC Langmuir monolayers as cell membrane models, Colloids and Surfaces B: Biointerfaces, vol.108, pp.120-126, 2013.

C. L. Tran, D. Buchanan, R. T. Cullen, A. Searl, J. Ad et al., Inhalation of poorly soluble particles. II. Influence Of particle surface area on inflammation and clearance, Inhalation toxicology, vol.12, pp.1113-1126, 2000.

B. Trouiller, R. Reliene, A. Westbrook, P. Solaimani, and R. H. Schiestl, Titanium dioxide nanoparticles induce DNA damage and genetic instability in vivo in mice, Cancer research, vol.69, pp.8784-8789, 2009.

J. Unnithan, M. U. Rehman, A. Fj, and M. Samim, Aqueous synthesis and concentration-dependent dermal toxicity of TiO2 nanoparticles in Wistar rats, Biological trace element research, vol.143, pp.1682-1694, 2011.

C. F. Urban, D. Ermert, M. Schmid, U. Abu-abed, C. Goosmann et al.,

, Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans, PLoS pathogens, vol.5, p.1000639

. Valverde and A. Castellanos, Fluidization, bubbling and jamming of nanoparticle agglomerates, Chemical Engineering Science, vol.62, pp.6947-6956, 2007.

P. M. Vanhoutte, Endothelial control of vasomotor function, Circulation journal, vol.67, pp.572-575, 2003.

. Veillenanos, Risques dioxyde de titane, Journal

K. V. Voelkerding, D. S. Durtschi, and J. D. , Next generation sequencing for clinical diagnostics-principles and application to targeted resequencing for hypertrophic cardiomyopathy, William Beaumont Hospital Symposium on Molecular Pathology. The Journal of molecular diagnostics : JMD, vol.12, pp.539-551, 2009.

N. Q. Vuong, P. Goegan, S. Mohottalage, D. Breznan, M. Ariganello et al., Proteomic changes in human lung epithelial cells (A549) in response to carbon black and titanium dioxide exposures, Journal of proteomics, vol.149, pp.53-63, 2016.

J. Wang, Y. Liu, F. Jiao, F. Lao, W. Li et al., Timedependent translocation and potential impairment on central nervous system by intranasally instilled TiO(2) nanoparticles, Toxicology, vol.254, pp.82-90, 2008.

W. Maldonado and M. A. , The ubiquitin-proteasome system and its role in inflammatory and autoimmune diseases, Cellular & molecular immunology, vol.3, pp.255-261, 2006.

J. Wang, G. Zhou, C. Chen, H. Yu, T. Wang et al., Acute toxicity and biodistribution of different sized titanium dioxide particles in mice after oral administration, Toxicology letters, vol.168, pp.176-185, 2007.

J. J. Wang, S. Bj, and H. Wang, Cyto-and genotoxicity of ultrafine TiO2 particles in cultured human lymphoblastoid cells, Mutation research, vol.628, pp.99-106, 2007.

Y. Wang, Z. Chen, T. Ba, J. Pu, T. Chen et al., Susceptibility of young and adult rats to the oral toxicity of titanium dioxide nanoparticles, Small, vol.9, pp.1742-1752, 2013.

Z. Wang, N. Li, J. Zhao, J. C. White, P. Qu et al., CuO nanoparticle interaction with human epithelial cells: cellular uptake, location, export, and genotoxicity, Chemical research in toxicology, vol.25, pp.1512-1521, 2012.

Z. Wang, C. Tiruppathi, J. Cho, and A. B. Malik, Delivery of nanoparticle: complexed drugs across the vascular endothelial barrier via caveolae, IUBMB life, vol.63, pp.659-667, 2011.

W. Pa and J. J. Grailer, Acute lung injury and the role of histones, Translational respiratory medicine, vol.2, p.1, 2014.

D. B. Warheit, How to measure hazards/risks following exposures to nanoscale or pigment-grade titanium dioxide particles, Toxicology letters, vol.220, pp.193-204, 2013.

D. B. Warheit, W. J. Brock, K. P. Lee, W. Tr, and K. L. Reed, Comparative pulmonary toxicity inhalation and instillation studies with different TiO2 particle formulations: impact of surface treatments on particle toxicity, Toxicological sciences : an official journal of the Society of Toxicology, vol.88, pp.514-524, 2005.

D. B. Warheit, B. Sc, and E. M. Donner, Acute and subchronic oral toxicity studies in rats with nanoscale and pigment grade titanium dioxide particles. Food and chemical toxicology : an international journal published for the, British Industrial Biological Research Association, vol.84, pp.208-224, 2015.

D. B. Warheit, J. F. Hansen, I. S. Yuen, D. P. Kelly, S. Si et al., Inhalation of high concentrations of low toxicity dusts in rats results in impaired pulmonary clearance mechanisms and persistent inflammation, Toxicology and applied pharmacology, vol.145, pp.10-22, 1997.

D. B. Warheit, H. Ma, and S. R. Frame, Pulmonary effects in rats inhaling size-separated chrysotile asbestos fibres or p-aramid fibrils: differences in cellular proliferative responses, Toxicology letters, vol.88, pp.287-292, 1996.

D. B. Warheit, R. A. Hoke, C. Finlay, E. M. Donner, R. Kl et al., Development of a base set of toxicity tests using ultrafine TiO2 particles as a component of nanoparticle risk management, Toxicology letters, vol.171, pp.99-110, 2007.

D. B. Warheit, T. R. Webb, K. L. Reed, S. Frerichs, and C. M. Sayes, Pulmonary toxicity study in rats with three forms of ultrafine-TiO2 particles: differential responses related to surface properties, Toxicology, vol.230, pp.90-104, 2007.

D. B. Warheit, I. S. Yuen, D. P. Kelly, S. Snajdr, and M. A. Hartsky, Subchronic inhalation of high concentrations of low toxicity, low solubility particulates produces sustained pulmonary inflammation and cellular proliferation, Toxicology letters, vol.88, pp.249-253, 1996.

. Waters and J. M. Fostel, Toxicogenomics and systems toxicology: aims and prospects, Nature reviews Genetics, vol.5, pp.936-948, 2004.

S. Wiese, K. A. Reidegeld, M. He, and B. Warscheid, Protein labeling by iTRAQ: a new tool for quantitative mass spectrometry in proteome research, Proteomics, vol.7, pp.340-350, 2007.

W. Ae and R. C. Chambers, The mercurial nature of neutrophils: still an enigma in ARDS?, American journal of physiology Lung cellular and molecular physiology, vol.306, pp.217-230, 2014.

J. Wong, M. Be, and L. J. Wood, Lung inflammation caused by inhaled toxicants: a review, International journal of chronic obstructive pulmonary disease, vol.11, pp.1391-1401, 2016.

J. Wu, W. Liu, C. Xue, S. Zhou, F. Lan et al., Toxicity and penetration of TiO2 nanoparticles in hairless mice and porcine skin after subchronic dermal exposure, Toxicology letters, vol.191, pp.1-8, 2009.

N. Wu, J. Wang, T. De, N. Wang, H. Zheng et al., Shapeenhanced photocatalytic activity of single-crystalline anatase TiO(2) (101) nanobelts, Journal of the American Chemical Society, vol.132, pp.6679-6685, 2010.

Y. Wu, Contact pathway of coagulation and inflammation, Thrombosis journal, vol.13, p.17, 2015.

J. Xu, Y. Sagawa, M. Futakuchi, K. Fukamachi, D. B. Alexander et al., Lack of promoting effect of titanium dioxide particles on ultraviolet B-initiated skin carcinogenesis in rats, Food and chemical toxicology : an international journal published for the British Industrial Biological Research Association, vol.49, pp.1298-1302, 2011.

M. Xu, L. Han, C. Yuan, Y. Mei, H. Zhang et al., Role for Functional SOD2 Polymorphism in Pulmonary Arterial Hypertension in a Chinese Population, p.14, 2017.

T. Xu, S. K. Park, J. D. Venable, J. A. Wohlschlegel, J. K. Diedrich et al., ProLuCID: An improved SEQUEST-like algorithm with enhanced sensitivity and specificity, Journal of proteomics, vol.129, pp.16-24

K. Yamashita, Y. Yoshioka, K. Higashisaka, K. Mimura, Y. Morishita et al.,

T. , Y. I. Saito, S. Tsutsumi, and Y. , Silica and titanium dioxide nanoparticles cause pregnancy complications in mice, Nature nanotechnology, vol.6, pp.321-328, 2011.

B. Yameen, W. I. Choi, C. Vilos, A. Swami, J. Shi et al., Insight into nanoparticle cellular uptake and intracellular targeting, Journal of controlled release : official journal of the Controlled Release Society, vol.190, pp.485-499, 2014.

B. Yang, Q. Wang, R. Lei, C. Wu, C. Shi et al., Systems toxicology used in nanotoxicology: mechanistic insights into the hepatotoxicity of nano-copper particles from toxicogenomics, Journal of nanoscience and nanotechnology, vol.10, pp.8527-8537, 2010.

X. Yu, H. F. Zhang, and Y. Q. , Bio-effect of nanoparticles in the cardiovascular system, Journal of Biomedical Materials Research Part A, vol.104, pp.2881-2897, 2016.

J. Zhang, P. Zhou, J. Liu, and Y. J. , New understanding of the difference of photocatalytic activity among anatase, rutile and brookite TiO2, Physical chemistry chemical physics : PCCP, vol.16, pp.20382-20386, 2014.

L. Zhang, M. Wang, X. Kang, P. Boontheung, N. Li et al., Oxidative stress and asthma: proteome analysis of chitinase-like proteins and FIZZ1 in lung tissue and bronchoalveolar lavage fluid, Journal of proteome research, vol.8, pp.1631-1638, 2009.

S. Zhang, H. Gao, and G. Bao, Physical Principles of Nanoparticle Cellular Endocytosis, ACS nano, vol.9, pp.8655-8671, 2015.

W. C. Zhang, X. J. Zheng, L. J. Du, J. Y. Sun, Z. X. Shen et al., High salt primes a specific activation state of macrophages, M(Na), Cell research, vol.25, pp.893-910, 2015.

, ANNEXE 2 : Morphologie, cytologie, histologie, fonction et structure de l'appareil respiratoire et régions utilisées dans le modèle dosimétrique de la Commission internationale de protection radiologique, 1994.

, Justification de l'exposition des rats à un aérosol nanostructuré de, ANNEXE, vol.3

, Aux Etats-Unis, le NIOSH a proposé en 2011 une VLEP pondérée sur 10h par jour et 40h par semaine de 0,3 mg/m 3 pour le TiO 2 ultrafin, 2011.

, En estimant une durée moyenne de vie au travail de 40 ans, avec environ 45 semaines travaillées par an, et 40h de travail par semaine

, Concernant certains paramètres respiratoires chez l'Homme : ? La fréquence respiratoire (Fr) représente la rapidité de respiration, Elle comprend environ, vol.12, issue.20

, ? Le volume courant VC (également appelé volume tidal) représente l'amplitude de respiration, c'est à dire le volume d'air inspiré ou expiré à chaque mouvement respiratoire. Au repos, le volume courant moyen est de 500 ml

, Ainsi le débit ventilatoire au repos est d'environ 6 à 10 L/min (= Fr x VC)

, En prenant la valeur par défaut du modèle MPPD de 7,5 L/min, le volume respiré tout au long de la carrière d'un Homme serait

L. Ainsi and . De, TiO 2 respirée au cours de la carrière d'un Homme serait, p.3

, En supposant que 10 % de la quantité respirée soit déposée dans les poumons chez l'Homme (modèle de la CIPR, 1994), cela correspondrait à une quantité déposée de 972 mg, mg/m 3 soit 9720 mg

, Cette quantité peut ensuite être normalisée par différents paramètres : ? Normalisation par poids corporel : En estimant le poids moyen d'un Homme à 70 kg et une quantité déposée de 972 mg (au cours de sa carrière), Pour un rat de poids moyen de 0,3 kg, cela correspond à une quantité déposée de 4,2 mg sur 1 mois

, En estimant la masse moyenne d'un poumon humain à 700 g et une quantité déposée de 972 mg (au cours de sa carrière)

, En estimant la surface alvéolaire totale moyenne des poumons chez l'Homme à 102 m 2 et une quantité déposée de 972 mg (au cours de sa carrière)

, Détermination de la concentration en aérosol de TiO 2 à utiliser par inhalation chez le rat

. En, 241 ml/min (Hsieh, 1999) ; le volume respiré sur 1 mois (6 h par jour, 5 jours par semaine, pendant 4 semaines) est de 1735,2 L soit environ 1,7 m 3 . Ainsi la concentration en aérosol (mg/m 3 ) : ? a été estimée à 9,6 mg/m 3 (environ 10 mg/m 3 ) en supposant 10 % de dépôt chez le rat et en tenant compte de la normalisation par masse de poumon : En effet, la quantité déposée au niveau pulmonaire est estimée à 1,7 mg sur 1 mois. Elle correspondant à une quantité respirée sur 1 mois de 17 mg

, mg/m 3 ) en se basant sur 20 % de dépôt chez le rat et une normalisation par surface alvéolaire : En effet, la quantité déposée au niveau alvéolaire est estimée à 3,8 mg sur 1 mois. Elle correspond à une quantité respirée sur 1 mois de 5 x 3,8 soit 19 mg

, Abbreviations: BALF, bronchoalveolar lavage fluid

. David, Database for Annotation, Visualization and Integrated Discovery; d0, d3 and d180, immediately, 3 and 180 post-exposure days respectively

, EBC, exhaled breath condensate; EMMS, Electro-Medical Measurement System; GO BP, gene ontology biological processes

, International Agency for Research on Cancer; KEGG, Kyoto Encyclopedia of Genes and Genomes

. Mudpit, Multidimensional Protein Identification Technology

. Nps and . Oecd, Organization for Economic Co-operation and Development; PBS, phosphate buffered saline

, ROS, reactive oxygen species; TiO2, titanium dioxide Abbreviations: BALF, bronchoalveolar lavage fluid

. David, Database for Annotation, Visualization and Integrated Discovery; d0, d3 and d180, immediately, 3 and 180 post-exposure days respectively

, EBC, exhaled breath condensate; EMMS, Electro-Medical Measurement System; GO BP, gene ontology biological processes

, International Agency for Research on Cancer; KEGG, Kyoto Encyclopedia of Genes and Genomes

. Mudpit, Multidimensional Protein Identification Technology

. Nps and . Oecd, Organization for Economic Co-operation and Development; PBS, phosphate buffered saline

, ROS