R. Das, B. F. Leo, and F. Murphy, The Toxic Truth About Carbon Nanotubes in Water Purification: 611 a Perspective View, Nanoscale Res. Lett, vol.13, p.183, 2018.

, Globe Newswire: Carbon Nanotubes Market By Product Type, Method And Application -613 Global Industry Analysis And Forecast To, 2022.

E. J. Petersen, L. Zhang, N. T. Mattison, D. M. O'carroll, A. J. Whelton et al.,

T. B. Huang, R. D. Henry, K. L. Holbrook, and . Chen, Potential Release Pathways, Environmental 616 Fate, And Ecological Risks of Carbon Nanotubes, Environ. Sci. Technol, vol.45, pp.9837-9856, 2011.

S. Kotsilkov, E. Ivanov, and N. K. Vitanov, Release of Graphene and Carbon Nanotubes from 618 Biodegradable Poly(Lactic Acid) Films during Degradation and Combustion: Risk Associated 619 with the End-of-Life of Nanocomposite Food Packaging Materials, Mater. Basel Switz, vol.11, p.620, 2018.

V. K. Upadhyayula, S. Deng, M. C. Mitchell, and G. B. Smith, Application of carbon nanotube 622 technology for removal of contaminants in drinking water: A review, Sci. Total Environ, vol.408, pp.1-13, 2009.

G. Chen, J. Qiu, Y. Liu, R. Jiang, S. Cai et al., Carbon 625 Nanotubes Act as Contaminant Carriers and Translocate within Plants, Sci. Rep, vol.5, p.15682, 2015.

M. Karimi, N. Solati, M. Amiri, H. Mirshekari, E. Mohamed et al.,

M. A. Saeidi, P. Estiar, A. Kiani, S. M. Ghasemi, A. R. Basri et al., Carbon 629 nanotubes part I: preparation of a novel and versatile drug-delivery vehicle

, Drug Deliv, vol.12, pp.1071-1087, 2015.

Y. Sun, K. Fu, Y. Lin, and W. Huang, Functionalized Carbon Nanotubes: Properties and 632 Applications, Acc. Chem. Res, vol.35, pp.1096-1104, 2002.

M. J. Rahman and T. Mieno, Conductive Cotton Textile from Safely Functionalized Carbon 634 Nanotubes, J. Nanomater, 2015.

Y. Hwang, S. Park, and J. W. Lee, Applications of Functionalized Carbon Nanotubes for the 636 Therapy and Diagnosis of Cancer, Polymers, vol.9, p.13, 2017.

M. Prato, K. Kostarelos, and A. Bianco, Functionalized carbon nanotubes in drug design and 638 discovery, Acc. Chem. Res, vol.41, pp.60-68, 2008.

S. Vardharajula, S. Z. Ali, P. M. Tiwari, E. Ero?lu, K. Vig et al., Functionalized 640 carbon nanotubes: biomedical applications, Int. J. Nanomedicine, vol.7, pp.5361-5374, 2012.

A. Yaghoubi and A. Ramazani, Synthesis of Amino-functionalized Carbon Nanotubes and 642 their Applications, Curr. Org. Chem, vol.22, pp.1505-1522, 2018.

L. Zhou, H. J. Forman, Y. Ge, and J. Lunec, Multi-walled carbon nanotubes: A cytotoxicity study in 644 relation to functionalization, dose and dispersion, Toxicol. In Vitro, vol.42, pp.292-298, 2017.

M. Jang and Y. S. Hwang, Effects of functionalized multi-walled carbon nanotubes on toxicity 646 and bioaccumulation of lead in Daphnia magna, PLoS ONE, vol.13, p.194935, 2018.

D. Gutiérrez-praena, S. Pichardo, E. Sánchez, A. Grilo, A. M. Cameán et al., Influence of 648 carboxylic acid functionalization on the cytotoxic effects induced by single wall carbon 649 nanotubes on human endothelial cells (HUVEC), Toxicol. Vitro Int. J. Publ. Assoc. BIBRA, vol.25, pp.1883-1888, 2011.

S. Jain, Toxicity Issues Related to Biomedical Applications of Carbon Nanotubes

, Nanomedicine Nanotechnol, vol.03, p.5, 2012.

A. Magrez, S. Kasas, V. Salicio, N. Pasquier, J. W. Seo et al.,

. Forró, Cellular Toxicity of Carbon-Based Nanomaterials, Nano Lett, vol.6, pp.1121-1125, 2006.

L. Zhou, Toxicity evaluation and medical application of multi-walled carbon nanotubes, p.656, 2015.

C. L. Ursini, D. Cavallo, A. M. Fresegna, A. Ciervo, R. Maiello et al., , p.658

S. Buresti and . Iavicoli, Study of Cytotoxic and Genotoxic Effects of Hydroxyl-Functionalized 659

, Multiwalled Carbon Nanotubes on Human Pulmonary Cells, J. Nanomater, 2012.

J. Azizian, H. Tahermansouri, E. Biazar, S. Heidari, and D. C. Khoei, Functionalization of 661 carboxylated multiwall nanotubes with imidazole derivatives and their toxicity 662 investigations, Int. J. Nanomedicine, vol.5, pp.907-914, 2010.

W. Chen, Q. Xiong, Q. Ren, Y. Guo, and G. Li, Can amino-functionalized carbon nanotubes carry 664 functional nerve growth factor?, Neural Regen. Res, vol.9, pp.285-292, 2014.

C. Buzea, I. I. Pacheco, and K. Robbie, Nanomaterials and nanoparticles: Sources and toxicity, vol.2, pp.17-71, 2007.

M. Wiemann, A. Vennemann, U. G. Sauer, K. Wiench, L. Ma-hock et al., An in vitro 668 alveolar macrophage assay for predicting the short-term inhalation toxicity of 669 nanomaterials, J. Nanobiotechnology, vol.14, p.16, 2016.

K. Fujita, M. Fukuda, S. Endoh, J. Maru, H. Kato et al.,

. Honda, Size effects of single-walled carbon nanotubes on in vivo and in vitro pulmonary 672 toxicity, Inhal. Toxicol, vol.27, pp.207-223, 2015.

K. Pulskamp, S. Diabaté, and H. F. Krug, Carbon nanotubes show no sign of acute toxicity but 674 induce intracellular reactive oxygen species in dependence on contaminants, Toxicol. Lett, vol.675, pp.58-74, 2007.

X. Kou, C. Li, D. He, X. Wang, T. Hao et al., Estradiol 677 Promotes M1-like Macrophage Activation through Cadherin-11 To Aggravate 678 Temporomandibular Joint Inflammation in Rats, J. Immunol, vol.194, pp.2810-2818, 2015.

C. Ronzani, R. Safar, R. Diab, J. Chevrier, J. Paoli et al., Joubert, Viability and gene expression responses to polymeric nanoparticles in human 681 and rat cells, Cell Biol. Toxicol, vol.680, pp.137-146, 2014.

P. Jackson, K. Kling, K. A. Jensen, P. A. Clausen, A. M. Madsen et al., 683 Characterization of genotoxic response to 15 multiwalled carbon nanotubes with variable 684 physicochemical properties including surface functionalizations in the FE1-Muta(TM) mouse 685 lung epithelial cell line: Functionalized MWCNT In Vitro, Environ. Mol. Mutagen, vol.56, pp.183-203, 2015.

S. Nahle, R. Safar, S. Grandemange, B. Foliguet, M. Lovera-leroux et al., Single wall and multiwall carbon nanotubes induce different 689 toxicological responses in rat alveolar macrophages, J. Appl. Toxicol

M. A. Ramakrishnan, Determination of 50% endpoint titer using a simple formula, World J

. Virol, , vol.5, pp.85-86, 2016.

H. Eidi, O. Joubert, C. Némos, S. Grandemange, B. Mograbi et al.,

A. L. Maincent, I. Faou, B. H. Aboukhamis, and . Rihn, Drug delivery by polymeric nanoparticles 694 induces autophagy in macrophages, Int. J. Pharm, vol.422, pp.495-503, 2012.

R. Safar, Z. Doumandji, T. Saidou, L. Ferrari, S. Nahle et al., Cytotoxicity and 696 global transcriptional responses induced by zinc oxide nanoparticles NM 110 in PMA-697 differentiated THP-1 cells, Toxicol. Lett, 2018.

A. Brazma, Minimum Information About a Microarray Experiment (MIAME) -Successes, p.699

, Failures, Challenges, Sci. World J, vol.9, pp.420-423, 2009.

T. Barrett and R. Edgar, Mining Microarray Data at NCBI\'s Gene Expression Omnibus (GEO) *, p.701

, Gene Mapp. Discov. Expr, pp.175-190, 2006.

Y. Benjamini and Y. Hochberg, Controlling the False Discovery Rate: A Practical and Powerful 703 Approach to Multiple Testing, J. R. Stat. Soc. Ser. B Methodol, vol.57, pp.289-300, 1995.

C. S. Hughes, S. Foehr, D. A. Garfield, E. E. Furlong, L. M. Steinmetz et al., Ultrasensitive 705 proteome analysis using paramagnetic bead technology, Mol. Syst. Biol, vol.10, pp.757-757, 2014.

C. Ramus, A. Hovasse, M. Marcellin, A. Hesse, E. Mouton-barbosa et al., , p.707

C. Carapito, K. Chaoui, C. Bruley, J. Garin, S. Cianférani et al., , p.708

C. Schiltz, Y. Schaeffer, A. Couté, and . Gonzalez-de-peredo, Spiked proteomic standard dataset for 709 testing label-free quantitative software and statistical methods, vol.6, pp.286-710, 2016.

J. Rappsilber, M. Mann, and Y. Ishihama, Protocol for micro-purification, enrichment, pre-712 fractionation and storage of peptides for proteomics using StageTips, Nat. Protoc, vol.2, pp.713-1896, 2007.

M. Allegri, D. K. Perivoliotis, M. G. Bianchi, M. Chiu, A. Pagliaro et al.,

E. Trompeta, O. Bergamaschi, C. A. Bussolati, and . Charitidis, Toxicity determinants of multi-716 walled carbon nanotubes: The relationship between functionalization and agglomeration, p.717

, Toxicol. Rep, vol.3, pp.230-243, 2016.

S. Y. Madani, A. Mandel, and A. M. Seifalian, A concise review of carbon nanotube's toxicology, 719 Nano Rev, vol.4, 2013.

I. Fenoglio, E. Aldieri, E. Gazzano, F. Cesano, M. Colonna et al.,

Y. Attanasio, D. Yakoub, B. Lison, and . Fubini, Thickness of Multiwalled Carbon Nanotubes Affects 722 Their Lung Toxicity, Chem. Res. Toxicol, vol.25, pp.74-82, 2012.

C. Loos, T. Syrovets, A. Musyanovych, V. Mailänder, K. Landfester et al., Amino-724 functionalized nanoparticles as inhibitors of mTOR and inducers of cell cycle arrest in 725 leukemia cells, Biomaterials, vol.35, pp.1944-1953, 2014.

M. Ema, H. Takehara, M. Naya, H. Kataura, K. Fujita et al., Length effects of single-727 walled carbon nanotubes on pulmonary toxicity after intratracheal instillation in rats

, Toxicol. Sci, vol.42, pp.367-378, 2017.

M. Rapp, M. W. Wintergerst, W. G. Kunz, V. K. Vetter, M. M. Knott et al.,

S. Haubner, R. Moder, S. Thaler, B. Eiber, N. Meyer et al.,

B. Layritz, S. Kühnemuth, C. Stutte, U. H. Bourquin, S. Von-andrian et al., CCL22 732 controls immunity by promoting regulatory T cell communication with dendritic cells in 733 lymph nodes, J. Exp. Med, vol.216, pp.1170-1181, 2019.

M. Pescatori, D. Bedognetti, E. Venturelli, C. Ménard-moyon, C. Bernardini et al.,

G. Piana, R. Maida, F. Manetti, A. Sgarrella, L. G. Bianco et al., Functionalized carbon 736 nanotubes as immunomodulator systems, Biomaterials, vol.34, pp.4395-4403, 2013.

O. Llorca, J. Martín-benito, M. Ritco-vonsovici, J. Grantham, G. M. Hynes et al., , p.738

J. M. Carrascosa and . Valpuesta, Eukaryotic chaperonin CCT stabilizes actin and tubulin folding 739 intermediates in open quasi-native conformations, EMBO J, vol.19, pp.5971-5979, 2000.

A. Panariti, G. Miserocchi, and I. Rivolta, The effect of nanoparticle uptake on cellular behavior: 741 disrupting or enabling functions?, Nanotechnol. Sci. Appl, vol.5, pp.87-100, 2012.

Y. Yao, E. Jones, and K. Inoki, Lysosomal Regulation of mTORC1 by Amino Acids in Mammalian 743 Cells, Biomolecules, 2017.

L. Bar-peled, L. D. Schweitzer, R. Zoncu, and D. M. Sabatini, Ragulator is a GEF for the rag GTPases 745 that signal amino acid levels to mTORC1, Cell, vol.150, pp.1196-1208, 2012.

V. Iadevaia, X. Wang, Z. Yao, L. J. Foster, and C. G. Proud, Evaluation of mTOR-regulated mRNA 747 translation, Methods Mol. Biol. Clifton NJ, vol.821, pp.171-185, 2012.

M. Lunova, A. Prokhorov, M. Jirsa, M. Hof, A. Ol?y?ska et al., Nanoparticle core stability and surface functionalization drive the mTOR 750 signaling pathway in hepatocellular cell lines, Sci. Rep, vol.7, p.16049, 2017.

G. Vales, L. Rubio, and R. Marcos, Genotoxic and cell-transformation effects of multi-walled 752 carbon nanotubes (MWCNT) following in vitro sub-chronic exposures, J. Hazard. Mater, vol.306, pp.193-202, 2016.

S. Bhattacharya, K. Srinivasan, S. Abdisalaam, F. Su, P. Raj et al.,

S. Wakeland, S. Ghose, A. Mukherjee, and . Asaithamby, RAD51 interconnects between DNA 756 replication, DNA repair and immunity, Nucleic Acids Res, vol.45, pp.4590-4605, 2017.

S. Difilippantonio, A. Celeste, M. J. Kruhlak, Y. Lee, M. J. Difilippantonio et al.,

P. J. Jackson, A. Mckinnon, and . Nussenzweig, Distinct domains in Nbs1 regulate irradiation-759 induced checkpoints and apoptosis, J. Exp. Med, vol.204, pp.1003-1011, 2007.

C. Deng, BRCA1: cell cycle checkpoint, genetic instability, DNA damage response and 761 cancer evolution, Nucleic Acids Res, vol.34, p.1416, 2006.

L. Feng, N. Li, Y. Li, J. Wang, M. Gao et al., Cell cycle-dependent inhibition of 763 53BP1 signaling by BRCA1, Cell Discov, vol.1, p.15019, 2015.

J. S. Kim, K. S. Song, and I. J. Yu, Multiwall Carbon Nanotube-Induced DNA Damage and 765 Cytotoxicity in Male Human Peripheral Blood Lymphocytes, Int. J. Toxicol, vol.35, pp.27-37, 2016.

A. Manke, L. Wang, and Y. , Mechanisms of Nanoparticle-Induced Oxidative Stress 767 and Toxicity, BioMed Res. Int, pp.1-15, 2013.

A. Patlolla, B. Knighten, and P. Tchounwou, Multi-Walled Carbon Nanotubes Induce Cytotoxicity, p.769

, Genotoxicity And Apoptosis In Normal Human Dermal Fibroblast Cells, vol.20, pp.1-65, 2010.

M. Luo, Z. Bao, F. Xu, X. Wang, F. Li et al., Unrepaired DNA damage 772 in macrophages causes elevation of particulate matter-induced airway inflammatory 773 response, Aging, vol.10, pp.549-560, 2018.

Y. Yang, G. Jiang, P. Zhang, and J. Fan, Programmed cell death and its role in inflammation, Mil

, Med. Res, vol.2, p.12, 2015.

Q. Yu, Y. V. Katlinskaya, C. J. Carbone, B. Zhao, K. V. Katlinski et al.,

T. Chen, C. J. Yang, R. A. Lengner, F. B. Greenberg, S. Y. Johnson et al., DNA-damage-induced 778 type I interferon promotes senescence and inhibits stem cell function, Cell Rep, vol.11, pp.785-797, 2015.

X. Hu and L. B. Ivashkiv, Cross-regulation of signaling pathways by interferon-gamma: 781 implications for immune responses and autoimmune diseases, Immunity, vol.31, pp.539-782, 2009.

W. Song, L. Popp, J. Yang, A. Kumar, V. S. Gangoli et al., The autophagic response to 784 polystyrene nanoparticles is mediated by transcription factor EB and depends on surface 785 charge, J. Nanobiotechnology, vol.13, 2015.

J. Harris, Autophagy and cytokines, Cytokine, vol.56, pp.140-144, 2011.

H. Liu, Y. Zhang, N. Yang, Y. Zhang, X. Liu et al.,

P. Zhang, F. Yang, Y. Guo, C. Sun, and . Jiang, A functionalized single-walled carbon nanotube-789 induced autophagic cell death in human lung cells through Akt-TSC2-mTOR signaling, Cell, vol.790, p.159

C. Pavan and B. Fubini, Unveiling the Variability of "Quartz Hazard" in Light of Recent 792 Toxicological Findings, Chem. Res. Toxicol, vol.30, pp.469-485, 2017.

A. Michihara, K. Toda, T. Kubo, Y. Fujiwara, K. Akasaki et al., Disruptive Effect of 794 Chloroquine on Lysosomes in Cultured Rat Hepatocytes, Biol. Pharm. Bull, vol.28, pp.947-795, 2005.

G. Thomas and M. N. Hall, TOR signalling and control of cell growth, Curr. Opin. Cell Biol, vol.9, pp.782-787, 1997.

H. B. Jefferies, S. Fumagalli, P. B. Dennis, C. Reinhard, R. B. Pearson et al., Rapamycin 799 suppresses 5'TOP mRNA translation through inhibition of p70s6k, EMBO J, vol.16, pp.3693-800, 1997.

T. Weichhart, G. Costantino, M. Poglitsch, M. Rosner, M. Zeyda et al., The TSC-mTOR 803 signaling pathway regulates the innate inflammatory response, Immunity, vol.29, pp.565-804, 2008.

H. R. Turnquist, J. Cardinal, C. Macedo, B. R. Rosborough, T. L. Sumpter et al., Metes, 806 A.W. Thomson, mTOR and GSK-3 shape the CD4+ T-cell stimulatory and differentiation 807 capacity of myeloid DCs after exposure to LPS, Blood, vol.115, pp.4758-4769, 2010.

F. O. Martinez and S. Gordon, The M1 and M2 paradigm of macrophage activation: time for 809 reassessment, 1000.

$. $-of$-carbon, $. Nanotubes$-in$-biotechnology, and $. Biomedicine,

, The$application$of$carbon$nanotubes$in$target$drug$delivery$systems$for$cancer$therapies

$. Available,

$. A. Bianco, $. K. Kostarelos, and $. M. Prato, Applications$of$carbon$nanotubes$in$drug$delivery, Curr.%Opin.%Chem.%Biol.,$, issue.9, pp.674-679, 2005.

, Mitochondria$ targeting$ nano$ agents$ in$ cancer$ therapeutics

, Carbon$ nanotubes$ as$ cancer$ therapeutic$ carriers$ and$ mediators

, NIOSH:$ Occupational$ Exposure$ to$ Carbon$ Nanotubes$ and$ Nanofibers$ |$ Nano

$. Y. Qin and $. , LongKterm$ intravenous$ administration$ of$ carboxylated$ singleKwalled$ carbon nanotubes$induces$persistent$accumulation$in$the$lungs$and$pulmonary$fibrosis$via$the$nuclear$factorK kappa$B$pathway, $Int.%J.%Nanomedicine,$, vol.12, pp.263-277, 2017.

$. L. Rahman$et%al, MultiKwalled$carbon$nanotubeKinduced$genotoxic,$inflammatory$and$proK fibrotic$responses$in$mice:$Investigating$the$mechanisms$of$pulmonary$carcinogenesis, $Mutat.%Res.,$, vol.823, pp.28-44, 2017.

$. Shvedova$et%al, Unusual$inflammatory$and$fibrogenic$pulmonary$responses$to$singleK walled$carbon$nanotubes$in$mice, $Am.%J.%Physiol.%Lung%Cell.%Mol.%Physiol.,$, vol.289, issue.5, 2005.

$. Shvedova$et%al, Inhalation$vs.$aspiration$of$singleKwalled$carbon$nanotubes$in$C57BL/6 mice:$ inflammation,$ fibrosis,$ oxidative$ stress,$ and$ mutagenesis, $ Am.% J.% Physiol.?Lung% Cell.% Mol. Physiol.,$, vol.295

$. R. Girardello, N. Baranzini, $. G. Tettamanti, $. M. De$eguileor, and $. A. Grimaldi, Cellular$responses$ induced$ by$ multiKwalled$ carbon$ nanotubes:$ in$ vivo$ and$ in$ vitro$ studies$ on$ the$ medicinal$ leech$ macrophages, $Sci.%Rep.,$, vol.8871, issue.1, 2017.

, The$toxic$effects$of$singleKwalled$carbon$nanotubes$are$linked$to$the$phagocytic$ability$of$cells K$ Toxicology$ Research$

, Mechanisms$ of$ lung$ fibrosis$ induced$ by$ carbon$ nanotubes:$ towards$ an$ Adverse$ Outcome$ Pathway$ (AOP)

K. , Size$effects$of$singleKwalled$carbon$nanotubes$on$in$vivo$and$in$vitro$pulmonary toxicity, $Inhal.%Toxicol.,$, vol.27, pp.207-223, 2015.

, MultiKwalled$carbon$nanotubes$induce$stronger$migration$of$inflammatory$cells$in$vitro$than$ asbestos$ or$ granular$ particles$ but$ a$ similar$ pattern$ of$ inflammatory$ mediators$ K$ ScienceDirect

%. S. Nahle%, Single% wall% and% multiwall% carbon% nanotubes% induce% different% toxicological responses%in%rat%alveolar%macrophages, %J.#Appl.#Toxicol
URL : https://hal.archives-ouvertes.fr/hal-01986763

, Elucidation%mechanism%of%different%biological%responses% to%multiDwalled%carbon%nanotubes using%four%cell%lines

, Biological%effects%of%doubleDwalled%carbon%nanotubes%on%the%innate%immune%system:%An%in% vitro% study% on% THPD1% human% monocytes.% D% PubMed% D% NCBI

%. J. %long, %. %ma, %. %yu, and %. , MultiDwalled%carbon%nanotubes%(MWCNTs)%promoted lipid%accumulation%in%THPD1%macrophages%through%modulation%of%endoplasmic%reticulum%(ER)%stress, Nanotoxicology

%. M. , Toxicity%determinants%of%multiDwalled%carbon%nanotubes:%The%relationship between%functionalization%and%agglomeration

, Complex% organic% corona% formation% on% carbon% nanotubes% reduces% microbial% toxicity% by% suppressing% reactive% oxygen% species% production% D% Environmental% Science:% Nano% (RSC% Publishing), p.13

%. A. %shvedova, %. %pietroiusti, %. , .. %. %kagan, and %. , Mechanisms%of%carbon%nanotubeD induced%toxicity:%Focus%on%oxidative%stress, 2012.

, Macrophage% sensing% of% singleDwalled% carbon% nanotubes% via% TollDlike% receptors% |% Scientific Reports, p.10

, Effect% of% Reconstituted% Pulmonary% Surfactant% Containing% the% 6000DDalton% Hydrophobic Protein%on%Lung%Compliance%of%Prematurely%Delivered%Rabbit%Fetuses%|%Pediatric%Research

, Barrier% or% carrier?% Pulmonary% surfactant% and% drug% delivery.% D% PubMed% D% NCBI

, Pulmonary%surfactant%and%nanocarriers:%Toxicity%versus%combined%nanomedical%applications.% D%PubMed%D%NCBI

%. R. Safar%, Cytotoxicity% and% global% transcriptional% responses% induced% by% zinc% oxide% nanoparticles%NM%110%in%PMADdifferentiated%THPD1%cells

%. W. %chanput, %. %. , .. %. %wichers, and %. , THPD1%cell%line:%An%in%vitro%cell%model%for%immune% modulation%approach

%. Academic,

%. G. Jia%, Cytotoxicity% of% Carbon Nanomaterials:% Single?Wall% Nanotube,% Multi?Wall Nanotube,%and%Fullerene

%. P. Kumarathasan%, Cytotoxicity% of% carbon% nanotube% variants:% A% comparative% in# vitro exposure% study%with%A549%epithelial%and%J774%macrophage% cells, % Nanotoxicology,%, vol.9

%. J. Knirsch, %. , .. %. %krug, and %. "oops%they%did%it%again!%carbon%nanotubes%-hoax%scientists%in%viability%assays,

%. Kim% and I. %. Yu, Single?Wall% Carbon% Nanotubes% (SWCNT)% Induce% Cytotoxicity% and Genotoxicity%Produced%by%Reactive%Oxygen%Species%(ROS)%Generation%in%Phytohemagglutinin%(PHA)? Stimulated%Male%Human%Peripheral%Blood%Lymphocytes,"%J.#Toxicol.#Environ.#Health#A,%vol.%77,%no.%19, pp.1141-1153

%. L. , Dispersion%of%single?walled%carbon%nanotubes%by%a%natural%lung%surfactant%for pulmonary%in%vitro%and%in%vivo%toxicity%studies

%. M. %rosenblatt, %. %. %burns, %. %e.%duncan, and .. %. %hughes,

%. R. %weissleder and %. %h.?c.%cheng, Magnetically%labeled%cells%can%be detected%by%MR%imaging, %J.#Magn.#Reson.#Imaging,%vol.%7,%no.%1,%pp.%258-263,%Jan.%1997.%

%. J. %sylvester, %. Ghodsian, %. %. , and .. %. %o'brien,

%. Y. %tsai%and%a and %. %m.%weissman, Ubiquitylation%in%ERAD: reversing%to%go%forward?, vol.9

%. J. %b.%durose, %. %scheuner, %. %j.%kaufman, and %. %. ,

%. X. %zhao, %. %chang, %. %long, %. %li, and %. , The%toxicity%of%multi?walled%carbon%nanotubes (MWCNTs)%to%human%endothelial%cells:%The%influence%of%diameters%of%MWCNTs, vol.126

%. L. Yvan and . Charvet%, Combined% deficiency% of% ABCA1% and% ABCG1% promotes% foam% cell% accumulation%and%accelerates%atherosclerosis%in%mice, %J.#Clin.#Invest

%. A. %graham and %. , Mitochondrial%function%is%involved%in%regulation%of%cholesterol efflux%to%apolipoprotein%(apo)A?

%. X. %ye, %. %li, %. %hou, %. %gao, %. %yang et al., Oncotarget

%. S. Sheng, %. Y. Kang, %. Y. Guo, %. Q. Pu, %. M. Cai et al., Overexpression% of% Sirt3% inhibits% lipid% accumulation%in%macrophages%through%mitochondrial%IDH2%deacetylation, %Int.#J.#Clin.#Exp.#Pathol

, Formation%of%Multinucleated%Giant%Cells%In%Vitro%Is%Dependent%on%the%Stage%of%Monocyte%to% Macrophage% Maturation% |% Blood% Journal

%. H. %takano and %. , Comparison%of%the%activities%of%multinucleated%boneSresorbing%giant%cells%derived from% CD14Spositive% cells% in% the% synovial% fluids% of% rheumatoid% arthritis% and% osteoarthritis% patients, % Rheumatology

%. A. Mcinnes, Interleukin% 4% induces% cultured% monocytes/macrophages% to% form% giant% multinucleated%cells, %J..Exp..Med

%. R. %endow, %. %w.%sullivan, and %. %. %carper, %and%G.%L.%Mandell,%"Induction%of%Multinucleated%Giant Cell% Formation% from% In. Vitro% Culture% of% Human% Monocytes% with% InterleukinS3% and% InterferonS?:% Comparison%with%Other%Stimulating%Factors

J. Respir, Cell.Mol..Biol

%. Quinn%-and%-i.%-a.%-schepetkin, Role% of% NADPH% Oxidase% in% Formation% and% Function% of% Multinucleated%Giant%Cells, %J..Innate.Immun

%. C. %mcclean%and%d, %M.%Tobin,%"Macrophage%form,%function,%and%phenotype%in%mycobacterial infection:%lessons%from%tuberculosis%and%other%diseases

. %-"foamy%macrophages%and%the%progression%of%the%human%tb%granuloma,

%. M. Sp and . Puissegur%et, Mycobacterial%Lipomannan%Induces%Granuloma%Macrophage%Fusion%via a% TLR2SDependent,% ADAM9S% and% ?1% IntegrinSMediated% Pathway, % J.. Immunol.,%, vol.178, issue.5, pp.3161-3169

, An%Overview%of%the%Derivation%and%Function%of%Multinucleated%Giant%Cells%and%Their%Role%in% Pathologic% Processes% S% The% American% Journal% of% Pathology

%. I. Iavicoli, %. %. Calabrese, and %. M. Nascarella, Exposure% to% Nanoparticles% and% Hormesis

%. F. %huaux%, %"ILS1?% induces% CD11b% low % alveolar%macrophage% proliferation%and%maturation during%granuloma%formation:%ILS1?%is%crucial%for%macrophage%proliferation, %J..Pathol

, Alveolar% macrophages% and% pulmonary% surfactant more% than% just% friendly% neighbours

%. J. %kay, %. %. %murray, %. %k.%pagan, .. %. %stow, and %. Raftsassociated%snares%at%the%phagocytic%cup, %J..Biol..Chem

%. Desai% and N. Leitinger, Purinergic% and% calcium% signaling% in% macrophage% function% and% plasticity

%. A. Ohradanova0repic%, Extracellular% Purine% Metabolism% Is% the% Switchboard% of% Immunosuppressive% Macrophages% and% a% Novel% Target% to% Treat% Diseases% With% Macrophage% Imbalances

%. M. %a.%samie%and%h.%xu and %. Lysosomal%exocytosis%and%lipid%storage%disorders, %J.#Lipid#Res

%. C. %t.%esmon and %. The%interactions%between%inflammation%and%coagulation,

%. Y. %kang, %. %a.%tan, %. %. %carroll, %. %. , and .. %. %sim, Target%pattern%recognition%by% complement%proteins%of%the% classical%and%alternative% pathways, % Adv.#Exp.#Med.#Biol.,%, vol.653, pp.117-128

%. J. Meng, %. M. Yang, %. F. Jia, %. Z. Xu, %. H. Kong et al., Immune% responses% of% BALB/c% mice% to subcutaneously%injected%multi0walled%carbon%nanotubes, 2011.

%. Ndika, %. J. Sund, %. H. Alenius, %. , and A. Puustinen, Elucidating% differential% nano0bio interactions% of% multi0walled% andsingle0walled% carbon% nanotubes% using% subcellular% proteomics, Nanotoxicology

%. S. %butcher and %. The%spliceosome%and%its%metal%ions, , 2011.

%. A. %j, Mapping%macrophage% polarization%over% the%myocardial%infarction%time% continuum

%. A. %berger, %. %h.%tran, .. %. %paige, and %. , Co0regulated%decrease% of%Neurokinin01%receptor%and Hemokinin01%gene%expression%in%monocytes%and%macrophages%after%activation%with%pro0inflammatory cytokines, %J.#Neuroimmunol

%. M. ,

%. M. %orecchioni, %. %ghosheh, and %. %. , Macrophage%Polarization:%Different%Gene% Signatures%in%M1(LPS+)%vs.%Classically%and%M2(LPS-)%vs.%Alternatively%Activated%Macrophages, %Front. Immunol

%. M. %fontana, %. %baccarella, %. %pancholi, %. %. %pufall, %. %. et al., JUNB%Is%a% Key%Transcriptional%Modulator%of%Macrophage%Activation, %J.#Immunol, 2015.

%. R. Shrivastava%, % "M2% polarization% of% macrophages% by% Oncostatin% M% in% hypoxic% tumor% microenvironment%is%mediated%by%mTORC2%and%promotes%tumor%growth%and%metastasis

%. J. , Serpin%2a%Is%Induced%in%Activated%Macrophages%and%Conjugates%to% a% Ubiquitin%Homolog, %J.#Immunol

, Revised proposal for the revision of the statistical definitions of biotechnology and nanotechnology, Technology and Industry Working Papers, 2018.

, Définition des nanomatériaux : Recommandations de la Commission européenne ANSM : Agence nationale de sécurité du médicament et des produits de santé

, Les nanomatériaux manufacturés à l'horizon 2030 en France Brochure INRS

E. Hood, Diving into the Unknown. Environmental Health Perspective, Nanotechnology, vol.112, 2004.

, Nanotechnology: A Realistic Market Assessment: NAN031F | BCC Research

A. Baezasquiban, Impacts physiopathologiques des nanoparticules inhalées, Biol. Aujourdhui, vol.208, issue.2, pp.151-158, 2014.

T. E. Allen, J. M. Goodman, S. Gutsell, and P. J. Russell, Defining Molecular Initiating Events in the Adverse Outcome Pathway Framework for Risk Assessment, Chem. Res. Toxicol, vol.27, issue.12, pp.2100-2112, 2014.

G. T. Ankley, Adverse outcome pathways: A conceptual framework to support ecotoxicology research and risk assessment, Environ. Toxicol. Chem, vol.29, issue.3, pp.730-741, 2010.

, Carbon black vs. black carbon and other airborne materials containing elemental carbon: Physical and chemical distinctions ScienceDirect

, Trends, and Forecasts, 2019.

, Nouveau procédé d'élaboration à grande échelle de films d'oxyde de graphène réduit

C. Han, Hypericinfunctionalized graphene oxide for enhanced mitochondriatargeting and synergistic anticancer effect, Acta Biomater, vol.77, pp.268-281, 2018.

S. Biswas and V. P. Torchilin, Nanopreparations for organellespecific delivery in cancer, Adv. Drug Deliv. Rev, vol.66, pp.26-41, 2014.

, Les nanotubes de carbone : quels risques, quelle prévention ? Article de revue INRS

, Engineering of oriented carbon nanotubes in composite materials

A. Eatemadi, Carbon nanotubes: properties, synthesis, purification, and medical applications, Nanoscale Res. Lett, vol.9, issue.1, p.393, 2014.

M. Kaseem, K. Hamad, and Y. G. Ko, Fabrication and materials properties of polystyrene/carbon nanotube (PS/CNT) composites: A review, Eur. Polym. J, vol.79, pp.36-62, 2016.

, Synthesis of carbon nanostructures by arc evaporation of Technische Informationsbibliothek (TIB)

, Mass production of singlewall carbon nanotubes by the arc plasma jet method ScienceDirect

, Carbon Nanotubes: Applications in Pharmacy and Medicine

, Carbon Nanotubes for Sensing Applications ScienceDirect

, Carbon nanotube membranes for water purification: Developments, challenges, and prospects for the future ScienceDirect

, PDF) Application potential of carbon nanotubes in water treatment: A review

L. Milane, M. Trivedi, A. Singh, M. Talekar, and M. Amiji, Mitochondrial biology, targets, and drug delivery, J. Controlled Release, vol.207, pp.40-58, 2015.

, Detection of cancer cells using a peptide nanotubefolic acid modified graphene electrode. PubMed NCBI

, Collagencarbon nanotube composite materials as scaffolds in tissue engineering. PubMed NCBI

, Carbon Nanotubes Market | Applications | Manufacturers | Size | Shares, DataMIntelligence

E. J. Petersen, Potential Release Pathways, Environmental Fate, And Ecological Risks of Carbon Nanotubes, Environ. Sci. Technol, vol.45, issue.23, pp.9837-9856, 2011.

V. L. Colvin, The potential environmental impact of engineered nanomaterials, Nat. Biotechnol, vol.21, issue.10, pp.1166-1170, 2003.

X. Zhao and R. Liu, Recent progress and perspectives on the toxicity of carbon nanotubes at organism, organ, cell, and biomacromolecule levels, Environ. Int, vol.40, pp.244-255, 2012.

, Les nanomatériaux. Définitions, risques toxicologiques, caractérisation de l'exposition professionnelle et mesures de prévention Brochure INRS

&. Paris, des nanotubes de carbone dans les poumons d'enfants asthmatiques

Z. Chen, Acute toxicological effects of copper nanoparticles in vivo, Toxicol. Lett, vol.163, issue.2, pp.109-120, 2006.

Z. Li, Cardiovascular effects of pulmonary exposure to singlewall carbon nanotubes, Environ. Health Perspect, vol.115, issue.3, pp.377-382, 2007.

L. Leclerc, Internalisation cellulaire et activité biologique de mico et nanoparticules fluorescentes de chimie de surface contrôlée, 2011.

P. Jackson, Bioaccumulation and ecotoxicity of carbon nanotubes, Chem. Cent. J, vol.7, issue.1, p.154, 2013.

B. A. Magnuson, T. S. Jonaitis, and J. W. Card, A Brief Review of the Occurrence, Use, and Safety of FoodRelated Nanomaterials, J. Food Sci, vol.76, issue.6, pp.126-133, 2011.

K. Kuche, Carbon nanotubes (CNTs) based advanced dermal therapeutics: current trends and future potential, Nanoscale, vol.10, issue.19, pp.8911-8937, 2018.

G. Liang, Effects of Subchronic Exposure to MultiWalled Carbon Nanotubes on Mice, J. Toxicol. Environ. Health A, vol.73, issue.7, pp.463-470, 2010.

R. R. Mercer, Extrapulmonary transport of MWCNT following inhalation exposure, Part. Fibre Toxicol, vol.10, issue.1, p.38, 2013.

G. Oberdörster, V. Castranova, B. Asgharian, and P. Sayre, Inhalation Exposure to Carbon Nanotubes (CNT) and Carbon Nanofibers (CNF): Methodology and Dosimetry, J. Toxicol. Environ. Health Part B, vol.18, issue.3-4, pp.121-212, 2015.

A. Gajewicz, Advancing risk assessment of engineered nanomaterials: application of computational approaches, Adv. Drug Deliv. Rev, vol.64, issue.15, pp.1663-1693, 2012.

, AVIS de l'Anses en réponse à la 'Consultation publique de la commission européenne relatif à la modification des annexes de REACh en vue de la prise en compte des nanomatériaux' | Anses Agence nationale de sécurité sanitaire de l'alimentation, de l'environnement et du travail

X. Cui, B. Wan, Y. Yang, X. Ren, and L. H. Guo, Length effects on the dynamic process of cellular uptake and exocytosis of singlewalled carbon nanotubes in murine macrophage cells, Sci. Rep, vol.7, issue.1, p.1518, 2017.

M. Allegri, Toxicity determinants of multiwalled carbon nanotubes: The relationship between functionalization and agglomeration, Toxicol. Rep, vol.3, pp.230-243, 2016.

R. Sturm, Nanotubes in the human respiratory tract -Deposition modeling, Z. Für Med. Phys, vol.25, issue.2, pp.135-145, 2015.

F. A. Murphy, LengthDependent Retention of Carbon Nanotubes in the Pleural Space of Mice Initiates Sustained Inflammation and Progressive Fibrosis on the Parietal Pleura, Am. J. Pathol, vol.178, issue.6, pp.2587-2600, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00605097

M. S. Boyles, Multiwalled carbon nanotube induced frustrated phagocytosis, cytotoxicity and proinflammatory conditions in macrophages are length dependent and greater than that of asbestos, Toxicol. In Vitro, vol.29, issue.7, pp.1513-1528, 2015.

D. Elgrabli, Toxicité et clairance pulmonaires des nanotubes de carbone, 2008.

M. Dörger, S. Münzing, A. M. Allmeling, K. Messmer, and F. Krombach, Differential Responses of Rat Alveolar and Peritoneal Macrophages to ManMade Vitreous Fibers in Vitro, Environ. Res, vol.85, issue.3, pp.207-214, 2001.

X. Shi, A. Dem-bussche, R. H. Hurt, A. B. Kane, and H. Gao, Cell entry of one dimensional nanomaterials occurs by tip recognition and rotation, Nat. Nanotechnol, vol.6, issue.11, pp.714-719, 2011.

D. Van-berlo, Apoptotic, inflammatory, and fibrogenic effects of two different types of multi walled carbon nanotubes in mouse lung, Arch. Toxicol, vol.88, issue.9, pp.1725-1737, 2014.

C. Bussy, Critical role of surface chemical modifications induced by length shortening on multi walled carbon nanotubesinduced toxicity, Part. Fibre Toxicol, vol.9, issue.1, p.46, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00761589

A. Peigney, C. Laurent, E. Flahaut, R. R. Bacsa, and A. Rousset, Specific surface area of carbon nanotubes and bundles of carbon nanotubes, Carbon, vol.39, issue.4, pp.507-514, 2001.
URL : https://hal.archives-ouvertes.fr/hal-01003709

, Surface area is the biologically most effective dose metric for acute nanoparticle toxicity in the lung ScienceDirect

A. Hirsch, Functionalization of SingleWalled Carbon Nanotubes, Angew. Chem. Int. Ed, vol.41, issue.11, pp.1853-1859, 2002.

T. Zhang, Surface modification of multiwall carbon nanotubes determines the proinflammatory outcome in macrophage, J. Hazard. Mater, vol.284, pp.73-82, 2015.

R. F. Hamilton, Z. Wu, S. Mitra, and A. Holian, The Effects of Varying Degree of MWCNT Carboxylation on Bioactivity in Various In Vivo and In Vitro Exposure Models, Int. J. Mol. Sci, vol.19, issue.2, 2018.

M. Nicoletti, C. Capodanno, C. Gambarotti, and E. Fasoli, Proteomic investigation on bio corona of functionalized multiwalled carbon nanotubes, Biochim. Biophys. Acta Gen. Subj, vol.1862, issue.10, pp.2293-2303, 2018.

T. Zhang, M. Tang, Y. Yao, Y. Ma, and Y. Pu, <p>MWCNT interactions with protein: surface induced changes in protein adsorption and the impact of protein corona on cellular uptake and cytotoxicity</p>, International Journal of Nanomedicine, 72019.

C. Kinnear, T. L. Moore, L. Rodriguezlorenzo, B. Rothenrutishauser, and A. Petrifink, Form Follows Function: Nanoparticle Shape and Its Implications for Nanomedicine, Chem. Rev, vol.117, issue.17, pp.11476-11521, 2017.

Z. Wang, F. Li, and A. Stein, Direct Synthesis of Shaped Carbon Nanoparticles with Ordered Cubic Mesostructure, Nano Lett, vol.7, issue.10, pp.3223-3226, 2007.

R. Bacchetta, N. Santo, I. Valenti, D. Maggioni, M. Longhi et al., Comparative toxicity of three differently shaped carbon nanomaterials on Daphnia magna : does a shape effect exist?, Nanotoxicology, vol.12, issue.3, pp.201-223, 2018.

S. E. Gratton, The effect of particle design on cellular internalization pathways, Proc. Natl. Acad. Sci, vol.105, issue.33, pp.11613-11618, 2008.

V. Cohignac, Carbon nanotubes, but not spherical nanoparticles, block autophagy by a shape related targeting of lysosomes in murine macrophages, Autophagy, vol.14, issue.8, pp.1323-1334, 2018.
URL : https://hal.archives-ouvertes.fr/cea-01823746

Z. Huang, N. Geyer, P. Werner, J. De-boor, and U. Gösele, MetalAssisted Chemical Etching of Silicon: A Review: In memory of Prof. Ulrich Gösele, Adv. Mater, vol.23, issue.2, pp.285-308, 2011.

J. Hua, M. G. Vijver, M. K. Richardson, F. Ahmad, and W. J. Peijnenburg, Particle specific toxic effects of differently shaped zinc oxide nanoparticles to zebrafish embryos ( Danio rerio ): Particle specific toxicity of differently shaped ZnO NPs, Environ. Toxicol. Chem, vol.33, issue.12, pp.2859-2868, 2014.

S. George, Surface Defects on PlateShaped Silver Nanoparticles Contribute to Its Hazard Potential in a Fish Gill Cell Line and Zebrafish Embryos, ACS Nano, vol.6, issue.5, pp.3745-3759, 2012.

K. Pulskamp, S. Diabaté, and H. F. Krug, Carbon nanotubes show no sign of acute toxicity but induce intracellular reactive oxygen species in dependence on contaminants, Toxicol. Lett, vol.168, issue.1, pp.58-74, 2007.

S. Y. Madani, A. Mandel, and A. M. Seifalian, A concise review of carbon nanotube's toxicology, Nano Rev, vol.4, 2013.

V. E. Kagan, Direct and indirect effects of single walled carbon nanotubes on RAW 264.7 macrophages: Role of iron, Toxicol. Lett, vol.165, issue.1, pp.88-100, 2006.

D. B. Warheit, T. R. Webb, K. L. Reed, S. Frerichs, and C. M. Sayes, Pulmonary toxicity study in rats with three forms of ultrafineTiO2 particles: Differential responses related to surface properties, Toxicology, vol.230, issue.1, pp.90-104, 2007.

W. J. Sandberg, Comparison of noncrystalline silica nanoparticles in IL1? release from macrophages, Part. Fibre Toxicol, vol.9, issue.1, p.32, 2012.

A. , In vitro toxicity of carbon nanotubes, nanographite and carbon black, similar impacts of acid functionalization, Toxicol. In Vitro, vol.30, issue.1, pp.476-485, 2015.

A. , Toxicité in vitro et propriétés physicochimiques de nanotubes de carbone, École nationale supérieure des mines, 2014.

A. J. Raghavendra, K. Fritz, S. Fu, J. M. Brown, R. Podila et al., Variations in biocorona formation related to defects in the structure of single walled carbon nanotubes and the hyperlipidemic disease state, Sci. Rep, vol.7, issue.1, p.8382, 2017.

J. Muller, Structural Defects Play a Major Role in the Acute Lung Toxicity of Multiwall Carbon Nanotubes: Toxicological Aspects, Chem. Res. Toxicol, vol.21, issue.9, pp.1698-1705, 2008.

I. Fenoglio, Structural Defects Play a Major Role in the Acute Lung Toxicity of Multiwall Carbon Nanotubes: Physicochemical Aspects, Chem. Res. Toxicol, vol.21, issue.9, pp.1690-1697, 2008.

D. W. Porter, Mouse pulmonary dose and time courseresponses induced by exposure to multi walled carbon nanotubes, Toxicology, vol.269, issue.2-3, pp.136-147, 2010.

J. R. Harkema, K. J. Nikula, and W. M. Haschek, Respiratory System, Fundamentals of Toxicologic Pathology, pp.351-393, 2018.

J. G. Teeguarden, P. M. Hinderliter, G. Orr, B. D. Thrall, and J. G. Pounds, Particokine tics In Vitro: Dosimetry Considerations for In Vitro Nanoparticle Toxicity Assessments, vol.95, pp.300-312, 2007.

A. G. Lenz, Efficient Bioactive Delivery of Aerosolized Drugs to Human Pulmonary Epithelial Cells Cultured in Air-Liquid Interface Conditions, Am. J. Respir. Cell Mol. Biol, vol.51, issue.4, pp.526-535, 2014.

S. Beyeler, Acute effects of multiwalled carbon nanotubes on primary bronchial epithelial cells from COPD patients, Nanotoxicology, vol.12, issue.7, pp.699-711, 2018.

S. Chortarea, H. Barosova, M. J. Clift, P. Wick, A. Petrifink et al., Human Asthmatic Bronchial Cells Are More Susceptible to Subchronic Repeated Exposures of Aerosolized Carbon Nanotubes At Occupationally Relevant Doses Than Healthy Cells, ACS Nano, vol.11, issue.8, pp.7615-7625, 2017.

A. A. Kapralov, Adsorption of Surfactant Lipids by SingleWalled Carbon Nanotubes in Mouse Lung upon Pharyngeal Aspiration, ACS Nano, vol.6, issue.5, pp.4147-4156, 2012.

L. Wang, Dispersion of singlewalled carbon nanotubes by a natural lung surfactant for pulmonary in vitro and in vivo toxicity studies, Part. Fibre Toxicol, vol.7, issue.1, p.31, 2010.

R. D. Brohi, Toxicity of Nanoparticles on the Reproductive System in Animal Models: A Review, Front. Pharmacol, vol.8, p.606, 2017.

A. Schaper, Countering health threats by chemicals with a potential terrorist backgroundcreating a rapid alert system for Europe, Eur. J. Intern. Med, vol.23, issue.2, pp.63-66, 2012.

L. Chezeau, Étude au niveau pulmonaire du profil d'expression de gènes et de protéines chez le rat exposé par inhalation à un aérosol de particules nanostructurées de dioxyde de titane, 2018.

E. Bermudez, LongTerm Pulmonary Responses of Three Laboratory Rodent Species to Subchronic Inhalation of Pigmentary Titanium Dioxide Particles, Toxicol. Sci, vol.70, issue.1, pp.86-97, 2002.

S. Bettini, Foodgrade TiO2 impairs intestinal and systemic immune homeostasis, initiates preneoplastic lesions and promotes aberrant crypt development in the rat colon, Sci. Rep, vol.7, issue.1, p.40373, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01508951

A. P. Francis and T. Devasena, Toxicity of carbon nanotubes: A review, Toxicol. Ind. Health, p.748233717747472, 2018.

N. Kobayashi, H. Izumi, and Y. Morimoto, Review of toxicity studies of carbon nanotubes, J. Occup. Health, vol.59, issue.5, pp.394-407, 2017.

A. Magrez, Cellular Toxicity of CarbonBased Nanomaterials, Nano Lett, vol.6, issue.6, pp.1121-1125, 2006.

, Toxicity of graphenefamily nanoparticles: a general review of the origins and mechanisms | Particle and Fibre Toxicology | Full Text

,

, TEL Thèses en ligne Réponse macrophagique à des nanoparticules manufacturées : effets de leurs caractéristiques physicochimiques sur l'autophagie

R. Gref, Stealth' coronacore nanoparticles surface modified by polyethylene glycol (PEG): influences of the corona (PEG chain length and surface density) and of the core composition on phagocytic uptake and plasma protein adsorption, Colloids Surf. B Biointerfaces, vol.18, issue.3-4, pp.301-313, 2000.

M. P. Monopoli, C. Åberg, A. Salvati, and K. A. Dawson, Biomolecular coronas provide the biological identity of nanosized materials, Nat. Nanotechnol, vol.7, issue.12, pp.779-786, 2012.

S. Wan, The 'Sweet' Side of the Protein Corona: Effects of Glycosylation on Nanoparticle-Cell Interactions, ACS Nano, vol.9, issue.2, pp.2157-2166, 2015.

X. Cai, Characterization of carbon nanotube protein corona by using quantitative proteomics, Nanomedicine Nanotechnol. Biol. Med, vol.9, issue.5, pp.583-593, 2013.

X. Zhao, D. Lu, F. Hao, and R. Liu, Exploring the diameter and surface dependent conformational changes in carbon nanotubeprotein corona and the related cytotoxicity, J. Hazard. Mater, vol.292, pp.98-107, 2015.

D. Khang, Y. K. Lee, E. J. Choi, T. J. Webster, and S. H. Kim, Effect of the protein corona on nanoparticles for&nbsp;modulating cytotoxicity and immunotoxicity, Int. J. Nanomedicine, p.97, 2014.

, Binding of blood proteins to carbon nanotubes reduces cytotoxicity | PNAS

, The effect of protein corona composition on the interaction of carbon nanotubes with human blood platelets. PubMed NCBI

, Effects of serum proteins on intracellular uptake and cytotoxicity of carbon nanoparticles ScienceDirect

, Protein CoronaMediated Mitigation of Cytotoxicity of Graphene Oxide | ACS Nano

,

, Hard corona composition and cellular toxicities of the graphene sheets ScienceDirect

R. H. Michell, MEASUREMENT OF RATES OF PHAGOCYTOSIS: The Use of Cellular Monolayers, J. Cell Biol, vol.40, issue.1, pp.216-224, 1969.

X. Cui, B. Wan, Y. Yang, X. Ren, and L. H. Guo, Length effects on the dynamic process of cellular uptake and exocytosis of singlewalled carbon nanotubes in murine macrophage cells, Sci. Rep, vol.7, issue.1, p.1518, 2017.

P. M. Costa, M. Bourgognon, J. T. Wang, and K. T. , Functionalised carbon nanotubes: From intracellular uptake and cellrelated toxicity to systemic brain delivery, J. Controlled Release, vol.241, pp.200-219, 2016.

B. Yameen, W. I. Choi, C. Vilos, A. Swami, J. Shi et al., Insight into nanoparticle cellular uptake and intracellular targeting, J. Controlled Release, vol.190, pp.485-499, 2014.

H. H. Gustafson, D. Holtcasper, D. W. Grainger, and H. Ghandehari, Nanoparticle uptake: The phagocyte problem, Nano Today, vol.10, issue.4, pp.487-510, 2015.

A. Schinwald and K. Donaldson, Use of backscatter electron signals to visualise cell/nanowires interactions in vitro and in vivo; frustrated phagocytosis of long fibres in macrophages and compartmentalisation in mesothelial cells in vivo, Part. Fibre Toxicol, vol.9, issue.1, p.34, 2012.

F. A. Murphy, A. Schinwald, C. A. Poland, and K. Donaldson, The mechanism of pleural inflammation by long carbon nanotubes: interaction of long fibres with macrophages stimulates them to amplify pro inflammatory responses in mesothelial cells, Part. Fibre Toxicol, vol.9, issue.1, p.8, 2012.

Y. Li and J. Cao, The impact of multiwalled carbon nanotubes (MWCNTs) on macrophages: contribution of MWCNT characteristics, Sci. China Life Sci, vol.61, issue.11, pp.1333-1351, 2018.

S. Baoukina, L. Monticelli, and D. P. Tieleman, Interaction of Pristine and Functionalized Carbon Nanotubes with Lipid Membranes, J. Phys. Chem. B, vol.117, issue.40, pp.12113-12123, 2013.

M. S. Lord, M. Foss, and F. Besenbacher, Influence of nanoscale surface topography on protein adsorption and cellular response, Nano Today, vol.5, issue.1, pp.66-78, 2010.

, Alveolar macrophage priming by intravenous administration of chitin particles, polymers of N acetyl Dglucosamine, in mice. PubMed NCBI

J. Liu and A. J. Hopfinger, Identification of Possible Sources of Nanotoxicity from Carbon Nanotubes Inserted into Membrane Bilayers Using Membrane Interaction Quantitative Structure?Activity Relationship Analysis, Chem. Res. Toxicol, vol.21, issue.2, pp.459-466, 2008.

M. L. Di-giorgio, S. D. Bucchianico, A. M. Ragnelli, P. Aimola, S. Santucci et al., Effects of single and multi walled carbon nanotubes on macrophages: Cyto and genotoxicity and electron microscopy, Mutat. Res. Toxicol. Environ. Mutagen, vol.722, issue.1, pp.20-31, 2011.

L. Lacerda, How do functionalized carbon nanotubes land on, bind to and pierce through model and plasma membranes, Nanoscale, vol.5, issue.21, p.10242, 2013.

, GrapheneInduced Pore Formation on Cell Membranes. PubMed NCBI

, Interactions of Graphene Oxide with Model Cell Membranes: Probing Nanoparticle Attachment and Lipid Bilayer Disruption | Langmuir

,

X. Ma, SingleWalled Carbon Nanotubes Alter Cytochrome c Electron Transfer and Modulate Mitochondrial Function, ACS Nano, vol.6, issue.12, pp.10486-10496, 2012.

N. Gao, Steering Carbon Nanotubes to Scavenger Receptor Recognition by Nanotube Surface Chemistry Modification Partially Alleviates NF?B Activation and Reduces Its Immunotoxicity, ACS Nano, vol.5, issue.6, pp.4581-4591, 2011.

B. L. Allen, Mechanistic Investigations of Horseradish Peroxidase Catalyzed Degradation of SingleWalled Carbon Nanotubes, J. Am. Chem. Soc, vol.131, issue.47, pp.17194-17205, 2009.

, Mitofusins Mfn1and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. PubMed NCBI

, Loss of OPA1 perturbates the mitochondrial inner membrane structure and integrity, leading to cytochrome c release and apoptosis. PubMed NCBI

, Cytotoxicité de nanoparticules polymériques, vecteurs de médicaments | cithefor.univ lorraine.fr

, Mitochondrial dysfunction, perturbations of mitochondrial dynamics and biogenesis involved in endothelial injury induced by silica nanoparticles ScienceDirect, p.7, 2019.

, Titanium Dioxide Nanoparticles Induce Mitochondrial Dynamic Imbalance and Damage in HT22 Cells

D. Chen, T. A. Stueckle, S. Luanpitpong, Y. Rojanasakul, Y. Lu et al., Gene expression profile of human lung epithelial cells chronically exposed to singlewalled carbon nanotubes, Nanoscale Res. Lett, vol.10, issue.1, p.12, 2015.

C. Xu, Q. Liu, H. Liu, C. Zhang, W. Shao et al., Toxicological assessment of multiwalled carbon nanotubes in vitro: potential mitochondria effects on male reproductive cells, Oncotarget, vol.7, issue.26, 2016.

H. A. Zeinabad, A. Zarrabian, A. A. Saboury, A. M. Alizadeh, and M. Falahati, Interaction of single and multi wall carbon nanotubes with the biological systems: tau protein and PC12 cells as targets, Sci. Rep, vol.6, issue.1, p.26508, 2016.

A. Burman, H. Tanjore, and T. S. Blackwell, Endoplasmic reticulum stress in pulmonary fibrosis, Matrix Biol, pp.355-365, 2018.

J. Long, W. Ma, Z. Yu, H. Liu, and Y. Cao, Multiwalled carbon nanotubes (MWCNTs) promoted lipid accumulation in THP1 macrophages through modulation of endoplasmic reticulum (ER) stress, Nanotoxicology, pp.1-14, 2019.

J. Yuan, H. Gao, J. Sui, H. Duan, W. N. Chen et al., Cytotoxicity Evaluation of Oxidized Single Walled Carbon Nanotubes and Graphene Oxide on Human Hepatoma HepG2 cells: An iTRAQCoupled 2D LCMS/MS Proteome Analysis, Toxicol. Sci, vol.126, issue.1, pp.149-161, 2012.

X. Zhao, S. Chang, J. Long, J. Li, X. Li et al., The toxicity of multiwalled carbon nanotubes (MWCNTs) to human endothelial cells: The influence of diameters of MWCNTs, Food Chem. Toxicol, vol.126, pp.169-177, 2019.

J. Long, Y. Xiao, L. Liu, and Y. Cao, The adverse vascular effects of multiwalled carbon nanotubes (MWCNTs) to human vein endothelial cells (HUVECs) in vitro: role of length of MWCNTs, J. Nanobiotechnology, vol.15, issue.1, p.80, 2017.

M. Orecchioni, D. Bedognetti, F. Sgarrella, F. M. Marincola, A. Bianco et al., Impact of carbon nanotubes and graphene on immune cells, J. Transl. Med, vol.12, issue.1, p.138, 2014.

M. Pescatori, Functionalized carbon nanotubes as immunomodulator systems, Biomaterials, vol.34, issue.18, pp.4395-4403, 2013.

X. Yang, Proteomic profiling of RAW264.7 macrophage cells exposed to graphene oxide: insights into acute cellular responses, Nanotoxicology, vol.13, issue.1, pp.35-49, 2019.

G. M. Hilton, Mapping differential cellular protein response of mouse alveolar epithelial cells to multiwalled carbon nanotubes as a function of atomic layer deposition coating, Nanotoxicology, vol.11, issue.3, pp.313-326, 2017.

&. Mtor-gene-genecards, |. Mtor-protein, and |. Mtor-antibody,

S. K. Sohaebuddin, P. T. Thevenot, D. Baker, J. W. Eaton, and L. Tang, Nanomaterial cytotoxicity is composition, size, and cell type dependent, Part. Fibre Toxicol, vol.7, issue.1, p.22, 2010.

T. Xia, M. Kovochich, M. Liong, J. I. Zink, and A. E. Nel, Cationic Polystyrene Nanosphere Toxicity Depends on CellSpecific Endocytic and Mitochondrial Injury Pathways, ACS Nano, vol.2, issue.1, pp.85-96, 2008.

B. Wan, Singlewalled carbon nanotubes and graphene oxides induce autophagosome accumulation and lysosome impairment in primarily cultured murine peritoneal macrophages, Toxicol. Lett, vol.221, issue.2, pp.118-127, 2013.

, Nanoparticles and Viruses as Mitophagy Inducers in Immune Cells | Housam Eidi | Request PDF

V. Cohignac, Carbon nanotubes, but not spherical nanoparticles, block autophagy by a shape related targeting of lysosomes in murine macrophages, Autophagy, vol.14, issue.8, pp.1323-1334, 2018.
URL : https://hal.archives-ouvertes.fr/cea-01823746

W. Zhu, Nanomechanical mechanism for lipid bilayer damage induced by carbon nanotubes confined in intracellular vesicles, Proc. Natl. Acad. Sci, vol.113, pp.12374-12379, 2016.

C. Bussy, Intracellular fate of carbon nanotubes inside murine macrophages: pHdependent detachment of iron catalyst nanoparticles, Part. Fibre Toxicol, vol.10, issue.1, p.24, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00840223

J. S. Kim, K. S. Song, and I. J. Yu, Multiwall Carbon NanotubeInduced DNA Damage and Cytotoxicity in Male Human Peripheral Blood Lymphocytes, Int. J. Toxicol, vol.35, issue.1, pp.27-37, 2016.

L. Migliore, Carbon nanotubes induce oxidative DNA damage in RAW 264.7 cells, Environ. Mol. Mutagen, p.NANA, 2010.

L. Zhu, D. W. Chang, L. Dai, and Y. Hong, DNA Damage Induced by Multiwalled Carbon Nanotubes in Mouse Embryonic Stem Cells, Nano Lett, vol.7, issue.12, pp.3592-3597, 2007.

K. Yamashita, Carbon Nanotubes Elicit DNA Damage and Inflammatory Response Relative to Their Size and Shape, Inflammation, vol.33, issue.4, pp.276-280, 2010.

C. J. Lu, Graphene oxide nanosheets induce DNA damage and activate the base excision repair (BER) signaling pathway both in vitro and in vivo, Chemosphere, vol.184, pp.795-805, 2017.

. Scihub, Comparative protein profile of human hepatoma HepG2 cells treated with graphene and singlewalled carbon nanotubes: An iTRAQcoupled 2D LC-MS/MS proteome analysis, Toxicology Letters, vol.207, issue.3

, Assessment of the toxic potential of graphene family nanomaterials

B. D. Holt, Altered Cell Mechanics from the Inside: Dispersed Single Wall Carbon Nanotubes Integrate with and Restructure Actin, J. Funct. Biomater, vol.3, issue.2, pp.398-417, 2012.

B. D. Holt, P. A. Short, A. D. Rape, Y. Wang, M. F. Islam et al., Carbon Nanotubes Reorganize Actin Structures in Cells and ex Vivo, ACS Nano, vol.4, issue.8, pp.4872-4878, 2010.

E. Witasp, A. A. Shvedova, V. E. Kagan, and B. Fadeel, Singlewalled carbon nanotubes impair human macrophage engulfment of apoptotic cell corpses, Inhal. Toxicol, vol.21, issue.sup1, pp.131-136, 2009.

W. Moller, Ultrafine Particles Cause Cytoskeletal Dysfunctions in Macrophages, Toxicol. Appl. Pharmacol, vol.182, issue.3, pp.197-207, 2002.

H. Moche, Utilisation du carbure de tungstènecobalt (WCCo) comme témoin positif génotoxique nanoparticulaire et étude de la génotoxicité de candidats nanovecteurs de médicaments, 2014.

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, issue.3, pp.233-278, 2014.

M. Pelin, Graphene and graphene oxide induce ROS production in human HaCaT s kin keratinocytes: the role of xanthine oxidase and NADH dehydrogenase, Nanoscale, vol.10, issue.25, pp.11820-11830, 2018.

A. A. Shvedova, A. Pietroiusti, B. Fadeel, and V. E. Kagan, Mechanisms of carbon nanotube induced toxicity: Focus on oxidative stress, Toxicol. Appl. Pharmacol, vol.261, issue.2, pp.121-133, 2012.

A. Manke, L. Wang, and Y. Rojanasakul, Mechanisms of Nanoparticle Induced Oxidative Stress and Toxicity, BioMed Res. Int, vol.2013, pp.1-15, 2013.

C. S. Sharma, SingleWalled Carbon Nanotubes Induces Oxidative Stress in Rat Lung Epithelial Cells, J. Nanosci. Nanotechnol, vol.7, issue.7, pp.2466-2472, 2007.

R. K. Srivastava, Multiwalled carbon nanotubes induce oxidative stress and apoptosis in human lung cancer cell lineA549, Nanotoxicology, vol.5, issue.2, pp.195-207, 2011.

K. Pulskamp, S. Diabate, and H. Krug, Carbon nanotubes show no sign of acute toxicity but induce intracellular reactive oxygen species in dependence on contaminants, Toxicol. Lett, vol.168, issue.1, pp.58-74, 2007.

V. E. Kagan, Carbon nanotubes degraded by neutrophil myeloperoxidase induce less pulmonary inflammation, Nat. Nanotechnol, vol.5, issue.5, pp.354-359, 2010.

M. Pacurari, Raw SingleWall Carbon Nanotubes Induce Oxidative Stress and Activate MAPKs, AP1, NF?B, and Akt in Normal and Malignant Human Mesothelial Cells, Environ. Health Perspect, vol.116, issue.9, pp.1211-1217, 2008.

L. Zhou, H. J. Forman, Y. Ge, and J. Lunec, Multiwalled carbon nanotubes: A cytotoxicity study in relation to functionalization, dose and dispersion, Toxicol. In Vitro, vol.42, pp.292-298, 2017.

Z. Tang, Mechanisms of oxidative stress, apoptosis, and autophagy involved in graphene oxide nanomaterial antiosteosarcoma effect, Int. J. Nanomedicine, vol.13, pp.2907-2919, 2018.

G. Qu, Graphene Oxide Induces Tolllike Receptor 4 (TLR4)Dependent Necrosis in Macrophages, ACS Nano, vol.7, issue.7, pp.5732-5745, 2013.

A. C. Reisetter, Induction of Inflammasomedependent Pyroptosis by Carbon Black Nanoparticles, J. Biol. Chem, vol.286, issue.24, pp.21844-21852, 2011.

S. Hussain, Inflammasome activation in airway epithelial cells after multiwalled carbon nanotube exposure mediates a profibrotic response in lung fibroblasts, Part. Fibre Toxicol, vol.11, issue.1, p.28, 2014.

V. Pongrakhananon, S. Luanpitpong, T. A. Stueckle, L. Wang, U. Nimmannit et al., Carbon Nanotubes Induce Apoptosis Resistance of Human Lung Epithelial Cells Through FLICEInhibitory Protein, Toxicol. Sci, vol.143, issue.2, pp.499-511, 2015.

J. Dong and Q. Ma, Advances in mechanisms and signaling pathways of carbon nanotube toxicity, Nanotoxicology, vol.9, issue.5, pp.658-676, 2015.

K. Lategan, H. Alghadi, M. Bayati, M. De-cortalezzi, and E. Pool, Effects of Graphene Oxide Nanoparticles on the Immune System Biomarkers Produced by RAW 264.7 and Human Whole Blood Cell Cultures, Nanomaterials, vol.8, issue.2, p.125, 2018.

A. C. Reisetter, Induction of Inflammasomedependent Pyroptosis by Carbon Black Nanoparticles, J. Biol. Chem, vol.286, issue.24, pp.21844-21852, 2011.

I. Fenoglio, Thickness of Multiwalled Carbon Nanotubes Affects Their Lung Toxicity, Chem. Res. Toxicol, vol.25, issue.1, pp.74-82, 2012.

S. Sweeney, D. Berhanu, S. K. Misra, A. J. Thorley, E. Valsamijones et al., Multiwalled carbon nanotube length as a critical determinant of bioreactivity with primary human pulmonary alveolar cells, Carbon, vol.78, pp.26-37, 2014.

J. Palomäki, A secretomics analysis reveals major differences in the macrophage responses towards different types of carbon nanotubes, Nanotoxicology, vol.9, issue.6, pp.719-728, 2015.

E. M. Rydman, Inhalation of rodlike carbon nanotubes causes unconventional allergic airway inflammation, Part. Fibre Toxicol, vol.11, p.48, 2014.

A. C. Grecco, Upregulation of T lymphocyte and antibody production by inflammatory cytokines released by macrophage exposure to multiwalled carbon nanotubes, Nanotechnology, vol.22, issue.26, p.265103, 2011.

S. Chen, No involvement of alveolar macrophages in the initiation of carbon nanoparticle induced acute lung inflammation in mice, Part. Fibre Toxicol, vol.13, issue.1, p.33, 2015.

C. Pavan and B. Fubini, Unveiling the Variability of 'Quartz Hazard' in Light of Recent Toxicological Findings, Chem. Res. Toxicol, vol.30, issue.1, pp.469-485, 2017.

B. Sun, NADPH OxidaseDependent NLRP3 Inflammasome Activation and its Important Role in Lung Fibrosis by Multiwalled Carbon Nanotubes, Small, vol.11, issue.17, pp.2087-2097, 2015.

R. F. Hamilton, Z. Wu, S. Mitra, P. K. Shaw, and A. Holian, Effect of MWCNT size, carboxylation, and purification on in vitro and in vivo toxicity, inflammation and lung pathology, Part. Fibre Toxicol, vol.10, issue.1, p.57, 2013.

S. P. Mukherjee, K. Kostarelos, and B. Fadeel, Cytokine Profiling of Primary Human Macrophages Exposed to EndotoxinFree Graphene Oxide: SizeIndependent NLRP3 Inflammasome Activation, Adv. Healthc. Mater, vol.7, issue.4, p.1700815, 2018.

M. Matsumoto, No toxicological effects on acute and repeated oral gavage doses of single wall or multiwall carbon nanotube in rats, J. Toxicol. Sci, vol.37, issue.3, pp.463-474, 2012.

G. Gerencsér, T. Varjas, K. Szendi, and C. Varga, In Vivo Induction of Primary DNA Lesions upon Subchronic Oral Exposure to Multiwalled Carbon Nanotubes, In Vivo, vol.30, issue.6, pp.863-868, 2016.

, Myofibroblasts and lung fibrosis induced by carbon nanotube ex posure | Particle and Fibre Toxicology | Full Text

,

, Induction of cancerassociated fibroblastlike cells by carbon nanotubes dictates its tumorigenicity | Scientific Reports

, Inflammation in the pleural cavity following injection of multiwalled carbon nanotubes is dependent on their characteristics and the presence of IL1 genes, Nanotoxicology, vol.12, issue.6

, , p.11, 2019.

M. M. Cartwright, The pulmonary inflammatory response to multiwalled carbon nanotubes is influenced by gender and glutathione synthesis, Redox Biol, vol.9, pp.264-275, 2016.

. Fibers-|-free and . Fulltext, The Significance and Insignificance of Carbon NanotubeInduced Inflammation | HTML

J. A. Bourdon, Carbon black nanoparticle instillation induces sustained inflammation and genotoxicity in mouse lung and liver, Part. Fibre Toxicol, vol.9, issue.1, p.5, 2012.

&. Pseudogene,

D. Anthony, SAA drives proinflammatory heterotypic macrophage differentiation in the lung via CSF1Rdependent signaling, FASEB J, vol.28, issue.9, pp.3867-3877, 2014.

R. D. Ye and L. Sun, Emerging functions of serum amyloid A in inflammation, J. Leukoc. Biol, vol.98, issue.6, pp.923-929, 2015.

J. Dong and Q. Ma, Type 2 Immune Mechanisms in Carbon Nanotube Induced Lung Fibrosis, Front. Immunol, vol.9, p.1120, 2018.

A. A. Shvedova, Unusual inflammatory and fibrogenic pulmonary responses to single walled carbon nanotubes in mice, Am. J. Physiol.Lung Cell. Mol. Physiol, vol.289, issue.5, pp.698-708, 2005.

J. Dong and Q. Ma, Macrophage polarization and activation at the interface of multiwalled carbon nanotubeinduced pulmonary inflammation and fibrosis, Nanotoxicology, vol.12, issue.2, pp.153-168, 2018.

K. Otsuka, Longterm polarization of alveolar macrophages to a profibrotic phenotype after inhalation exposure to multiwall carbon nanotubes, PLOS ONE, vol.13, issue.10, p.205702, 2018.

M. F. Cesta, Bacterial Lipopolysaccharide Enhances PDGF Signaling and Pulmonary Fibrosis in Rats Exposed to Carbon Nanotubes, Am. J. Respir. Cell Mol. Biol, vol.43, issue.2, pp.142-151, 2010.

J. Dong and Q. Ma, Advances in mechanisms and signaling pathways of carbon nanotube toxicity, Nanotoxicology, vol.9, issue.5, pp.658-676, 2015.

H. Nagai, Diameter and rigidity of multiwalled carbon nanotubes are critical factors in mesothelial injury and carcinogenesis, Proc. Natl. Acad. Sci, vol.108, issue.49, pp.1330-1338, 2011.

K. Donaldson, F. A. Murphy, R. Duffin, and C. A. Poland, Asbestos, carbon nanotubes and the pleural mesothelium: a review and the hypothesis regarding the role of long fibre retention in the parietal pleura, inflammation and mesothelioma, Part. Fibre Toxicol, vol.7, issue.1, p.5, 2010.

S. Rittinghausen, The carcinogenic effect of various multiwalled carbon nanotubes (MWCNTs) after intraperitoneal injection in rats, Part. Fibre Toxicol, vol.11, issue.1, p.59, 2014.

, Évaluation des risques liés aux nanomatériaux | Anses Agence nationale de sécurité sanitaire de l'alimentation, de l'environnement et du travail

, Etude de la biodistribution et de la toxicité des Nanoparticules de Fer chez le rat et sur une lignée de neuroblastome, theses.fr -Dalel Askri

, Toxicité des nanoparticules et des nanotubes | Sciences et Dé mocratie

, Nanomaterials in REACH and CLP Environment European Commission

&. Reach and . Info,

, NIOSH: Occupational Exposure to Carbon Nanotubes and Nanofibers | Nano

, Carbon Nanotubes Exposure Risk Assessment: From Toxicology to Epidemiologic Studies (Overview of the Current Problem)

, Current intelligence bulletin 65: occupational exposure to carbon nanotubes and nanofibers, 20042517.

, Occupational Exposure Limits of lead, dimethylamine, nbutyl2,3epoxypropyl ether, and 2ethyl1 hexanol and carcinogenicity and occupational sensitizer classification

, Long Term and Standard Incubations of WST1 Reagent Reflect the Same Inhibitory Trend of Cell Viability in Rat Airway Smooth Muscle Cells

, Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessme nt of mammalian cell cytotoxicity. PubMed NCBI

F. K. Chan, K. Moriwaki, and M. J. De-rosa, Detection of necrosis by release of lactate dehydrogenase activity, Methods Mol. Biol. Clifton NJ, vol.979, pp.65-70, 2013.

, Singlepot, solidphaseenhanced sample preparation for proteomics experiments | Nature Protocols

M. Zhang and J. Li, Carbon nanotube in different shapes, Mater. Today, vol.12, issue.6, pp.12-18, 2009.

Y. Hiraku, Nitrative DNA damage induced by carbonblack nanoparticles in macrophages and lung epithelial cells, Mutat. Res. Toxicol. Environ. Mutagen, vol.818, pp.7-16, 2017.

J. Ma, From the lung to the knee joint: Toxicity evaluation of carbon black nanoparticles on macrophages and chondrocytes, J. Hazard. Mater, vol.353, pp.329-339, 2018.

S. Chortarea, Profibrotic Activity of Multiwalled Carbon Nanotubes Upon Prolonged Exposures in Different Human Lung Cell Types, Appl. Vitro Toxicol, vol.5, issue.1, pp.47-61, 2019.

R. Bacchetta, N. Santo, I. Valenti, D. Maggioni, M. Longhi et al., Comparative toxicity of three differently shaped carbon nanomaterials on Daphnia magna : does a shape effect exist?, Nanotoxicology, vol.12, issue.3, pp.201-223, 2018.

A. M. Elgazzar, Comparative pulmonary toxicity of a DWCNT and MWCNT7 in rats, Arch. Toxicol, vol.93, issue.1, pp.49-59, 2019.

S. S. Poulsen, MWCNTs of different physicochemical properties cause similar inflammatory responses, but differences in transcriptional and histological markers of fibrosis in mouse lungs, Toxicol. Appl. Pharmacol, vol.284, issue.1, pp.16-32, 2015.

, In vitro study of human alveolar macrophages inflammatory mediator transcriptions and releases induced by soot FR 101, Printex 90, titandioxide and

J. C. Simard, F. Vallieres, R. De-liz, V. Lavastre, and D. Girard, Silver Nanoparticles Induce Degradation of the Endoplasmic Reticulum Stress Sensor Activating Transcription Factor6 Leading to Activation of the NLRP3 Inflammasome, J. Biol. Chem, vol.290, issue.9, pp.5926-5939, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01352655

A. Irachetavellve, Endoplasmic Reticulum Stressinduced Hepatocellular Death Pathways Mediate Liver Injury and Fibrosis via Stimulator of Interferon Genes, J. Biol. Chem, vol.291, issue.52, pp.26794-26805, 2016.

E. Borkhamkamphorst, E. Van-de-leur, U. Haas, and R. Weiskirchen, Liver parenchymal cells lacking Lipocalin 2 (LCN2) are prone to endoplasmic reticulum stress and unfolded protein response, Cell. Signal, vol.55, pp.90-99, 2019.

M. Polimeni, Multiwalled carbon nanotubes directly induce epithelialmesenchymal transition in human bronchial epithelial cells via the TGF?mediated Akt/GSK3?/SNAIL1 signalling pathway, Part. Fibre Toxicol, vol.13, issue.1, p.27, 2015.

S. Chortarea, Profibrotic Activity of Multiwalled Carbon Nanotubes Upon Prolonged Exposures in Different Human Lung Cell Types, Appl. Vitro Toxicol, vol.5, issue.1, pp.47-61, 2019.

H. Bian, MAPK/p38 regulation of cytoskeleton rearrangement accelerates induction of macrophage activation by TLR4, but not TLR3, Int. J. Mol. Med, vol.40, issue.5, pp.1495-1503, 2017.

P. T. Parthasarathy, Y. Cho, R. Lockey, and N. Kolliputi, An Old Molecule with a New Role: Microtubules in Inflammasome Regulation, Cell Biochem. Biophys, vol.70, issue.1, pp.697-698, 2014.

S. Carta, Histone deacetylase inhibitors prevent exocytosis of interleukin1betacontaining secretory lysosomes: role of microtubules, Blood, vol.108, issue.5, pp.1618-1626, 2006.

X. Zhu, Microtubules Negatively Regulate Insulin Secretion in Pancreatic ? Cells, Dev. Cell, vol.34, issue.6, pp.656-668, 2015.

P. Arenasguerrero, Determination of the size distribution of nonspherical nanoparticles by electric birefringencebased methods, Sci. Rep, vol.8, 2018.

T. Liu and Z. Xiao, Dynamic Light Scattering of Rigid Rods A Universal Relationship on the Apparent Diffusion Coefficient as Revealed by Numerical Studies and Its Use for Rod Length Determination, Macromol. Chem. Phys, vol.213, issue.16, pp.1697-1705, 2012.

B. N. Khlebtsov and N. G. Khlebtsov, On the measurement of gold nanoparticle sizes by the dynamic light scattering method, Colloid J, vol.73, issue.1, pp.118-127, 2011.

A. Fraczek, E. Menaszek, C. Paluszkiewicz, and M. Blazewicz, Comparative in vivo biocompatibility study of single and multiwall carbon nanotubes, Acta Biomater, vol.4, issue.6, pp.1593-1602, 2008.

C. Liné, C. Larue, and E. Flahaut, Carbon nanotubes: Impacts and behaviour in the terrestrial ecosystem A review, Carbon, vol.123, pp.767-785, 2017.

R. Liu, T. Gong, K. Zhang, and C. Lee, Graphene oxide papers with high water adsorption capacity for air dehumidification, Sci. Rep, vol.7, issue.1, p.9761, 2017.

A. Liang, X. Jiang, X. Hong, Y. Jiang, Z. Shao et al., Recent Developments Concerning the Dispersion Methods and Mechanisms of Graphene, Coatings, vol.8, issue.1, p.33, 2018.

, Different cellular response mechanisms contribute to the lengthdependent cytotoxicity of multi walled carbon nanotubes

M. S. Boyles, Multiwalled carbon nanotube induced frustrated phagocytosis, cytotoxicity and proinflammatory conditions in macrophages are length dependent and greater than that of asbestos, Toxicol. Vitro Int. J. Publ. Assoc. BIBRA, vol.29, issue.7, pp.1513-1528, 2015.

S. Lanone, P. Andujar, A. Kermanizadeh, and J. Boczkowski, Determinants of carbon nanotube toxicity, Adv. Drug Deliv. Rev, vol.65, issue.15, pp.2063-2069, 2013.

M. Landry, Early signs of multiwalled carbon nanotbues degradation in macrophages, via an intracellular pHdependent biological mechanism; importance of length and functionalization, Part. Fibre Toxicol, vol.13, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01401938

V. E. Kagan, Lung Macrophages 'Digest' Carbon Nanotubes Using a Superoxide/Peroxynitrite Oxidative Pathway, ACS Nano, vol.8, issue.6, pp.5610-5621, 2014.

, Functionalization density dependence of singlewalled carbon nanotubes cytotoxicity in vitro ScienceDirect

, In vitro toxicity of acidfunctionalized singlewalled carbon nanotubes: effects on murine macrophages and gene expression profiling. PubMed NCBI

S. Vardharajula, Functionalized carbon nanotubes: biomedical applications, Int. J. Nanomedicine, vol.7, pp.5361-5374, 2012.

, Pulmonary effects induced by ultrafine PTFE particles. PubMed NCBI

, Graphene oxide and reduced graphene oxide induced neural pheochromocyt | IJN

, Reduction of graphene oxide alters its cytocompatibility towards primary and immortalized macrophages Nanoscale

, Macrophage sensing of singlewalled carbon nanotubes via Tolllike receptors | Scientific Reports

J. Meng, X. Li, C. Wang, H. Guo, J. Liu et al., Carbon nanotubes activate macrophages into a M1/M2 mixed status: recruiting naïve macrophages and supporting angiogenesis, ACS Appl. Mater. Interfaces, vol.7, issue.5, pp.3180-3188, 2015.

, Disruption of protein homeostasis: a potential biomarker of exposure to three metal oxide nanoparticles ScienceDirect

R. Safar, Cytotoxicity and global transcriptional responses induced by zinc oxide nanoparticles NM 110 in PMAdifferentiated THP1 cells, Toxicol. Lett, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01952869

C. Xu, Q. Liu, H. Liu, C. Zhang, W. Shao et al., Toxicological assessment of multiwalled carbon nanotubes in vitro: potential mitochondria effects on male reproductive cells, Oncotarget, vol.7, issue.26, pp.39270-39278, 2016.

, Physical consequences of the mitochondrial targeting of singlewalled carbon nanotubes probed computationally ScienceDirect

, Toxicity of nano zinc oxide to mitochondria Toxicology Research

, AKAP13 Gene GeneCards | AKP13 Protein | AKP13 Antibody

, C12orf43 Gene GeneCards | CSTOS Protein | CSTOS Antibody

&. Snrnp27-gene-genecards, |. Snr27-protein, and |. Antibody,

&. Stambpl1-gene-genecards, |. Stalp-protein, and |. Stalp-antibody,

&. Genecards, |. Zc12a-protein, and |. Antibody,

C. Ge, The contributions of metal impurities and tube structure to the toxicity of carbon nanotube materials, NPG Asia Mater, vol.4, issue.12, p.32, 2012.

Z. Doumandji, T. Saidou, L. Ferrari, S. Nahle, B. Rihn et al., 2.1. Chemicals DMEM medium (Dulbecco's Modified Eagle's Medium-high glucose, D1145), fetal bovine serum (F7524), L-glutamine, Annexe 3. Cytotoxicity and global transcriptional responses induced by zinc oxide nanoparticles NM 110 in PMAdifferentiated THP1 cells. Ramia Safar, pp.150849-52

, AlamarBlue ® Cell Viability Reagent (DAL1025) was from Invitrogen (Villebon sur Yvette, France)

. Phuyal, sonicated using a 3-mm probe (Vibracell 75022, Bioblock, Illkirch, France) at 30% magnitude for 6 min under continuous cooling with ice. Immediately after sonication, ZnO110NP suspension was physico-chemically characterized and working dilutions in the culture medium were prepared. The hydrodynamic diameter and size distribution of ZnO110NP suspension, expressed as polydispersity index (PdI), were measured using dynamic light scattering (DLS, Zetasizer ? 3000E, Malvern Instruments Worcestershire, UK). Zeta potential was calculated using the Smoluchowski's equation (Sze et al., 2003). culture THP-1 human monocytic cell line was obtained from American Type Culture Collection, Preparation and characterization of nanoparticle suspension ZnO110NP were obtained from the Joint Research Center (NM 110, JRC), 2017.

. Ng/ml, nM) of PMA in a plate appropriate to the test for 24 h. The density of 5 × 10 4 cells per mL was respected for all tests. 2.4. Cell phenotype study After differentiation

. Thermo-fisher, After 5 min of centrifugation at 400 x g, cell pellet was resuspended in PBS-BSA. 4 × 10 5 cells were incubated with fluorescein isothiocyanate (FITC)-labeled monoclonal antibodies against CD11c for 30 min, then washed with PBS-BSA, and were analyzed by flow cytometry (BD Biosciences, THP-1 cells were treated with and without PMA in 6 wells UpCell plate

, Cytotoxicity study After treatment with PMA for 24 h, the medium was removed by using WST-1, LDH and, AlamarBlue ® assays. Unexposed cells were used as control and considered as having 100% of cell viability. Six wells and four test replicates were used per culture condition. The inhibitory concentration (IC 50 ) was calculated with the Reed and Muench method, 1938.

. Ronzani, Briefly, after 24 h of exposure to ZnO110NP, cells were incubated with 5% WST-1 reagent for 2 h at 37°C. Then, the absorbance was read at 450 nm with 690 nm as reference, 2.5.1. WST-1 assay WST-1 (water soluble tetrazolium) assay was performed as previously described, 2014.

, LDH assay Lactate dehydrogenase (LDH) assay was performed according to manufacturer's instructions. Briefly, after 24 h of exposure to ZnO110NP, cells were incubated with 100 ?L of (LDH reaction buffer + substrate) for 30 min at 25°C, Then, 50 ?L of stop solution was added and the absorbance was read at 490 nm (iMarkTM Microplate Absorbance Reader

, AlamarBlue ® assay was performed according to manufacturer's instructions. Briefly, cells were incubated with 10% of reagent for 3 h at 37°C. Fluorescence was read at 590 nm after excitation at 560 nm using spectrofluorimetry (JASCO, FMP-825

. Eidi, total RNA was, then, extracted using RNA-Solv Reagent ® according to manufacturer's protocols. The quality of the extracted RNA was assessed by spectrophotometry (BioSpecnano, Shimadzu) and capillary electrophoresis using RNA 6000 Nano ® (2100 Bioanalyzer?, Agilent Technologies, 2012.

R. Safar, Microarray hybridation cDNA and Cy3-dye-labeled cRNA synthesis were carried out with 100 ng of total RNA according to the manufacturer protocol (One-Color Microarray-Based Gene Expression Analysis, version 6.6). Then, 600 ng, Toxicology Letters, vol.308, pp.65-73, 2019.

R. Bauman, J. W. Liu, J. Klaassen, and C. D. , Production of metallothionein and heat-shock proteins in response to metals, Fundam. Appl. Toxicol, vol.21, issue.1, pp.15-22, 1993.

T. Buerki-thurnherr, L. Xiao, L. Diener, O. Arslan, C. Hirsch et al., In vitro mechanistic study towards a better understanding of ZnO nanoparticle toxicity, Nanotoxicology, vol.7, issue.4, pp.402-416, 2013.

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

B. De-berardis, G. Civitelli, M. Condello, P. Lista, R. Pozzi et al., Exposure to ZnO nanoparticles induces oxidative stress and cytotoxicity in human colon carcinoma cells, Toxicol. Appl. Pharmacol, vol.246, issue.3, pp.116-127, 2010.

H. Eidi, O. Joubert, C. Némos, S. Grandemange, B. Mograbi et al., Drug delivery by polymeric nanoparticles induces autophagy in macrophages, Int. J. Pharm, vol.422, issue.1-2, pp.495-503, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00746557

L. Farcal, F. Torres-andón, L. Di-cristo, B. M. Rotoli, O. Bussolati et al., Comprehensive in vitro toxicity testing of a panel of representative oxide nanomaterials: first steps towards an intelligent testing strategy, PLoS One, vol.10, issue.5, 2015.

F. Gao, N. Ma, H. Zhou, Q. Wang, H. Zhang et al., Zinc oxide nanoparticles-induced epigenetic change and G2/M arrest are associated with apoptosis in human epidermal keratinocytes, Int. J. Nanomed, vol.11, pp.3859-3874, 2016.

K. Girigoswami, Toxicity of metal oxide nanoparticles, Adv. Exp. Med. Biol, vol.122, pp.1048-1099, 2018.

G. H. Gwak, W. J. Lee, S. M. Paek, and J. M. Oh, Physico-chemical changes of ZnO nanoparticles with different size and surface chemistry under physiological pH conditions, Colloids Surf. B Biointerfaces, vol.127, pp.137-142, 2015.

C. Hanley, J. Layne, A. Punnoose, K. M. Reddy, I. Coombs et al., Preferential killing of cancer cells and activated human T cells using ZnO nanoparticles, Nanotechnology, vol.19, issue.29, p.295103, 2008.

C. Hanley, A. Thurber, C. Hanna, A. Punnoose, J. Zhang et al., The influences of cell type and ZnO nanoparticle size on immune cell cytotoxicity and cytokine induction, Nanoscale Res. Lett, vol.4, issue.12, pp.1409-1420, 2009.

A. Hartwig, Zinc finger proteins as potential targets for toxic metal ions: differential effects on structure and function, Antioxid. Redox Signal, vol.3, issue.4, pp.625-634, 2001.

W. Huang-da, B. T. Sherman, and R. A. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nat. Protoc, vol.4, issue.1, pp.44-57, 2009.

R. Hussien, B. H. Rihn, H. Eidi, C. Ronzani, O. Joubert et al., Unique growth pattern of human mammary epithelial cells induced by polymeric nanoparticles, Physiol. Rep, vol.422, issue.1-2, pp.495-503, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01272753

C. F. Jones and D. W. Grainger, In vitro assessments of nanomaterial toxicity, Adv. Drug Deliv. Rev, vol.61, issue.6, pp.438-456, 2009.

S. H. Joo and D. Zhao, Environmental dynamics of metal oxide nanoparticles in heterogeneous systems: a review, J. Hazard. Mater, vol.322, pp.29-47, 2017.

H. Liang, T. He, J. Long, L. Liu, G. Liao et al., Influence of bovine serum albumin pre-incubation on toxicity and ER stress-apoptosis gene expression in THP-1 macrophages exposed to ZnO nanoparticles, Toxicol. Mech. Methods, vol.21, pp.1-12, 2018.

P. K. Mishra, H. Mishra, A. Ekielski, S. Talegaonkar, and B. Vaidya, Zinc oxide nanoparticles: a promising nanomaterial for biomedical applications, Drug Discov. Today, vol.22, issue.12, pp.1825-1834, 2017.

P. J. Moos, K. Olszewski, M. Honeggar, P. Cassidy, S. Leachman et al., Responses of human cells to ZnO nanoparticles: a gene transcription study, Metallomics, vol.3, issue.11, pp.1199-1211, 2011.

S. Phuyal, M. Kasem, L. Rubio, H. L. Karlsson, R. Marcos et al., Effects on human bronchial epithelial cells following low-dose chronic exposure to nanomaterials: a 6-month transformation study, Toxicol. In Vitro, vol.44, pp.230-240, 2017.

M. Prach, V. Stone, and L. Proudfoot, Zinc oxide nanoparticles and monocytes: impact of size, charge and solubility on activation status, Toxicol. Appl. Pharmacol, vol.266, issue.1, pp.19-26, 2013.

M. Premanathan, K. Karthikeyan, K. Jeyasubramanian, and G. Manivannan, Selective toxicity of ZnO nanoparticles toward gram-positive bacteria and cancer cells by apoptosis through lipid peroxidation, Nanomedicine, vol.7, issue.2, pp.184-192, 2011.

L. J. Reed and H. Muench, A simple method of estimating fifty percent endpoints, Am. J. Hyg, vol.27, pp.493-497, 1938.

B. H. Rihn and O. Joubert, Protein corona fingerprinting predicts the cellular interaction of gold and silver nanoparticles, ACS Nano, vol.9, issue.6, pp.5634-5635, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01281135

C. Ronzani, R. Safar, R. Diab, J. Chevrier, J. Paoli et al., Viability and gene expression responses to polymeric nanoparticles in human and rat cells, Cell Biol. Toxicol, vol.30, issue.3, pp.137-146, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01274268

R. Roy, V. Parashar, L. K. Chauhan, R. Shanker, M. Das et al., Mechanism of uptake of ZnO nanoparticles and inflammatory responses in macrophages require PI3K mediated MAPKs signaling, Toxicol. In Vitro, vol.28, issue.3, pp.457-467, 2014.

R. Safar, C. Ronzani, R. Diab, J. Chevrier, D. Bensoussan et al., Human monocyte response to S-nitrosoglutathioneloaded nanoparticles: uptake, viability, and transcriptome, Mol. Pharm, vol.12, issue.2, pp.554-561, 2015.

D. Sahu, G. M. Kannan, and R. Vijayaraghavan, Size-dependent effect of zinc oxide on toxicity and inflammatory potential of human monocytes, J. Toxicol. Environ. Health A, vol.77, issue.4, pp.177-191, 2014.

V. A. Senapati, A. Kumar, G. S. Gupta, A. K. Pandey, and A. Dhawan, ZnO nanoparticles induced inflammatory response and genotoxicity in human blood cells: amechanistic approach, Food Chem. Toxicol, vol.85, pp.61-70, 2015.

A. Sze, D. Erickson, L. Ren, and D. Li, Zeta-potential measurement using the Smoluchowski equation and the slope of the current-time relationship in electroosmotic flow, J. Colloid Interface Sci, vol.261, issue.2, pp.402-410, 2003.

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 Res, vol.43, pp.447-452, 2015.

S. Tuomela, R. Autio, T. Buerki-thurnherr, O. Arslan, A. Kunzmann et al., Gene expression profiling of immune-competent human cells exposed to engineered zinc oxide or titanium dioxide nanoparticles, PLoS One, vol.8, issue.7, 2013.

M. E. Vance, T. Kuiken, E. P. Vejerano, S. P. Mcginnis, M. F. Hochella et al., Nanotechnology in the real world: redeveloping the nanomaterial consumer products inventory, Beilstein J. Nanotechnol, vol.21, issue.6, pp.1769-1780, 2015.

B. Wang, J. Zhang, C. Chen, G. Xu, X. Qin et al., The size of zinc oxide nanoparticles controls its toxicity through impairing autophagic flux in A549 lung epithelial cells, Toxicol. Lett, vol.285, pp.51-59, 2018.

V. Wilhelmi, U. Fischer, H. Weighardt, K. Schulze-osthoff, C. Nickel et al., Zinc oxide nanoparticles induce necrosis and apoptosis in macrophages in a p47phox-and Nrf2-independent manner, PLoS One, vol.8, issue.6, 2013.

H. Yin, P. S. Casey, M. J. Mccall, and M. Fenech, Size-dependent cytotoxicity and genotoxicity of ZnO particles to human lymphoblastoid (WIL2-NS) cells, Environ. Mol. Mutagen, vol.56, issue.9, pp.767-776, 2015.

R. Safar, Toxicology Letters, vol.308, pp.65-73, 2019.

H. A. Alhadlaq, M. J. Akhtar, and M. Ahamed, Zinc ferrite nanoparticle-induced 24 cytotoxicity and oxidative stress in different human cells, Cell Biosci, vol.5, p.55, 2015.

,

C. Andreini, L. Banci, I. Bertini, and A. Rosato, Counting the zinc-proteins encoded in 3 the human genome, J. Proteome Res, vol.5, pp.196-201, 2006.

C. Buzea, I. I. Pacheco, and K. Robbie, Nanomaterials and nanoparticles: sources and 5 toxicity, Biointerphases, vol.2, pp.17-71, 2007.

W. Chai, S. S. Zakrzewski, D. Günzel, R. Pieper, Z. Wang et al.,

N. Osterrieder and M. Burwinkel, High-dose dietary zinc oxide mitigates infection 8 with transmissible gastroenteritis virus in piglets, BMC Vet. Res, vol.10, p.75, 2014.

,

G. Chen, Y. Shen, X. Li, Q. Jiang, S. Cheng et al., The 11 endoplasmic reticulum stress inducer thapsigargin enhances the toxicity of ZnO 12 nanoparticles to macrophages and macrophage-endothelial co-culture, Environ. Toxicol, 2017.

. Pharmacol, , vol.50, pp.103-110

M. Chevallet, B. Gallet, A. Fuchs, P. H. Jouneau, K. Um et al., Metal homeostasis disruption and mitochondrial dysfunction in hepatocytes 16 exposed to sub-toxic doses of zinc oxide nanoparticles, Nanoscale, vol.15, pp.18495-18506, 2016.

,

C. C. Chusuei, C. Wu, S. Mallavarapu, F. Y. Hou, C. Hsu et al., , p.19

R. S. Aronstam and Y. Huang, Cytotoxicity in the age of nano: The role of fourth 20 period transition metal oxide nanoparticle physicochemical properties, Chem. Biol, 2013.

, , vol.206, pp.319-326

J. Cohen, G. Deloid, G. Pyrgiotakis, and P. Demokritou, Interactions of engineered 23 nanomaterials in physiological media and implications for in vitro dosimetry, 2013.

, Nanotoxicology, vol.7, pp.417-448

J. Cox, M. Y. Hein, C. A. Luber, I. Paron, N. Nagaraj et al., Accurate Proteome-1 wide Label-free Quantification by Delayed Normalization and Maximal Peptide Ratio, 2014.

, Extraction, Termed MaxLFQ. Mol. Cell. Proteomics, vol.13, pp.2513-2526

,

H. Eidi, O. Joubert, C. N??mos, S. Grandemange, B. Mograbi et al., , vol.5

P. Maincent, A. Le-faou, I. Aboukhamis, and B. H. Rihn, Drug delivery by 6 polymeric nanoparticles induces autophagy in macrophages, Int. J. Pharm, vol.422, pp.495-503, 2012.

,

E. Figueira, D. Branco, S. C. Antunes, F. Gonçalves, and R. Freitas, Are metallothioneins 9 equally good biomarkers of metal and oxidative stress?, Ecotoxicol. Environ. Saf, vol.84, pp.185-190, 2012.

K. Ganguly, D. Ettehadieh, S. Upadhyay, S. Takenaka, T. Adler et al., , p.12

W. G. Kreyling, H. Schulz, O. Schmid, and T. Stoeger, Early pulmonary response is 13 critical for extra-pulmonary carbon nanoparticle mediated effects: comparison of 14 inhalation versus intra-arterial infusion exposures in mice, Part. Fibre Toxicol, vol.14, p.19, 2017.

,

L. Gaté, C. Disdier, F. Cosnier, F. Gagnaire, J. Devoy et al., , p.17

M. Mabondzo and A. , Biopersistence and translocation to extrapulmonary organs of 18 titanium dioxide nanoparticles after subacute inhalation exposure to aerosol in adult and 19 elderly rats, Toxicol. Lett, vol.265, pp.61-69, 2017.

J. George and N. Ahmad, Mitochondrial Sirtuins in Cancer: Emerging Roles and 21, 2016.

, Therapeutic Potential, Cancer Res, vol.76, pp.2500-2506

L. Gesquière, N. Loreau, A. Minnich, J. Davignon, and D. Blache, Oxidative stress leads 24 to cholesterol accumulation in vascular smooth muscle cells. Free Radic, Biol. Med, vol.27, pp.134-179, 1999.

Y. Gong, Y. Ji, F. Liu, J. Li, and Y. Cao, Cytotoxicity, oxidative stress and inflammation 2 induced by ZnO nanoparticles in endothelial cells: interaction with palmitate or 3 lipopolysaccharide, J. Appl. Toxicol, vol.37, pp.895-901, 2017.

C. S. Hughes, S. Foehr, D. A. Garfield, E. E. Furlong, L. M. Steinmetz et al., , 2014.

, Ultrasensitive proteome analysis using paramagnetic bead technology, Mol. Syst. Biol, vol.6, p.757

S. Hussain, J. A. Vanoirbeek, and P. H. Hoet, Interactions of nanomaterials with the 8 immune system, Nanomedicine Nanobiotechnology, vol.4, pp.169-183, 2012.

,

P. Jennings, Stress response pathways, toxicity pathways and adverse outcome 11 pathways, Arch. Toxicol, vol.87, pp.13-17, 2013.

Y. Li, J. Bao, Y. Bian, U. Erben, P. Wang et al., , 2018.

, S100A4+ Macrophages Are Necessary for Pulmonary Fibrosis by Activating Lung 14

, Fibroblasts. Front. Immunol, vol.9, 1776.

J. Z. Lindeque, O. Levanets, R. Louw, and F. H. Van-der-westhuizen, The involvement of 16 metallothioneins in mitochondrial function and disease, Curr. Protein Pept. Sci, vol.11, pp.292-309, 2010.

I. Lynch and K. A. Dawson, Protein-nanoparticle interactions, Nano Today, vol.3, pp.40-47, 2008.

, , pp.70014-70022

M. Maiorino, M. Conrad, and F. Ursini, GPx4, Lipid Peroxidation, and Cell Death, p.21, 2018.

R. Discoveries and O. Issues, Antioxid. Redox Signal, vol.29, pp.61-74

,

. Mv-berridge, . Tan, and R. W. Mccoy, The biochemical and cellular basis of cell 24 proliferation assays that use tetrazolium salts, vol.4, pp.15-19, 1996.

K. Niska, K. Pyszka, C. Tukaj, M. Wozniak, M. W. Radomski et al., Titanium dioxide nanoparticles enhance production of superoxide anion and alter 2 the antioxidant system in human osteoblast cells, Int. J. Nanomedicine, vol.10, pp.1095-107, 2015.

,

G. Oberdörster, E. Oberdörster, and J. Oberdörster, Nanotoxicology: An Emerging, 2005.

, Discipline Evolving from Studies of Ultrafine Particles, Environ. Health Perspect, vol.113, pp.823-839

M. Prach, V. Stone, and L. Proudfoot, Zinc oxide nanoparticles and monocytes: impact of 8 size, charge and solubility on activation status, Toxicol. Appl. Pharmacol, vol.266, pp.19-26, 2013.

,

M. Présumé, A. Simon-deckers, C. Tomkiewicz-raulet, B. Le-grand, and J. Tran-van-nhieu, , p.11

G. Beaune, O. Duruphty, J. Doucet, X. Coumoul, J. Pairon et al., , p.12

S. Lanone and P. Andujar, Exposure to metal oxide nanoparticles administered at 13 occupationally relevant doses induces pulmonary effects in mice, Nanotoxicology, vol.10, pp.1535-1544, 2016.

L. J. Reed and H. Muench, A SIMPLE METHOD OF ESTIMATING FIFTY PER 16 CENT ENDPOINTS12, Am. J. Epidemiol, vol.27, pp.493-497, 1938.

,

R. Safar, Z. Doumandji, T. Saidou, L. Ferrari, S. Nahle et al., , 2018.

, Cytotoxicity and global transcriptional responses induced by zinc oxide nanoparticles 20 NM 110 in PMA-differentiated THP-1 cells, Toxicol. Lett

,

M. Sato and M. Kondoh, Recent Studies on Metallothionein: Protection Against 23 Toxicity of Heavy Metals and Oxygen Free Radicals, Tohoku J. Exp. Med, vol.196, pp.9-22, 2002.

,

R. P. Schins, J. H. Lightbody, P. J. Borm, T. Shi, K. Donaldson et al., , 2004.

, Inflammatory effects of coarse and fine particulate matter in relation to chemical and 2 biological constituents, Toxicol. Appl. Pharmacol, vol.195, pp.1-11

,

R. Singh and J. W. Lillard, Nanoparticle-based targeted drug delivery, Exp. Mol. Pathol, 2009.

, , vol.86, pp.215-223

E. Sozen and N. K. Ozer, Impact of high cholesterol and endoplasmic reticulum stress on 7 metabolic diseases: An updated mini-review, Redox Biol, vol.12, pp.456-461, 2017.

,

S. Tada-oikawa, G. Ichihara, Y. Suzuki, K. Izuoka, W. Wu et al., , p.10

S. Ichihara, Zn(II) released from zinc oxide nano/micro particles suppresses 11 vasculogenesis in human endothelial colony-forming cells, Toxicol. Reports, vol.2, pp.692-701, 2015.

,

J. Tannenbaum and B. T. Bennett, Russell and Burch's 3Rs then and now: the need for 14 clarity in definition and purpose, J. Am. Assoc. Lab. Anim. Sci, vol.54, pp.120-152, 2015.

T. J. Brunner, ?. , P. Wick, §. , P. Manser et al., , p.16

L. K. Limbach, ?. , A. Bruinink, J. Wendelin, *. Stark et al., , p.17, 2006.

, Cytotoxicity of Oxide Nanoparticles: Comparison to Asbestos, Silica, and the Effect of 18

, Particle Solubility ?

S. Tuomela, R. Autio, T. Buerki-thurnherr, O. Arslan, and A. Kunzmann,

B. Willman, P. Wick, S. Mathur, A. Scheynius, H. F. Krug et al., Gene Expression Profiling of Immune-Competent Human Cells Exposed, p.22, 2013.

, Engineered Zinc Oxide or Titanium Dioxide Nanoparticles, PLoS One, vol.8, p.68415

,

B. L. Vallee, The function of metallothionein, Neurochem. Int, vol.27, pp.23-33, 1995.

M. Va?ák, Advances in metallothionein structure and functions, J. Trace Elem. Med, 2005.

, Biol, vol.19, pp.13-17

H. Wallin, Z. O. Kyjovska, S. S. Poulsen, N. R. Jacobsen, A. T. Saber et al., , p.3

P. Vogel and U. , Surface modification does not influence the genotoxic and 4 inflammatory effects of TiO2 nanoparticles after pulmonary exposure by instillation in 5 mice, Mutagenesis, vol.32, pp.47-57, 2017.

M. Wiemann, A. Vennemann, U. G. Sauer, K. Wiench, L. Ma-hock et al., , 2016.

, An in vitro alveolar macrophage assay for predicting the short-term inhalation toxicity of 8 nanomaterials, J. Nanobiotechnology, vol.14

P. Yang, W. Hong, P. Zhou, B. Chen, and H. Xu, Nano and bulk ZnO trigger diverse Zn-10 transport-related gene transcription in distinct regions of the small intestine in mice after 11 oral exposure, Biochem. Biophys. Res. Commun, vol.493, pp.1364-1369, 2017.

,

X. Yang, H. Shao, W. Liu, W. Gu, X. Shu et al., , 2015.

, Endoplasmic reticulum stress and oxidative stress are involved in ZnO nanoparticle-15 induced hepatotoxicity, Toxicol. Lett, vol.234, pp.40-49

,

W. Zhang, S. Ohno, B. Steer, S. Klee, C. A. Staab-weijnitz et al., , vol.18

T. Stoeger, M. Königshoff, and H. Adler, S100a4 Is Secreted by Alternatively 19, 2018.

, Activated Alveolar Macrophages and Promotes Activation of Lung Fibroblasts in 20

, Pulmonary Fibrosis. Front. Immunol, vol.9, p.1216

,