S. Kaluza, J. Balderhaar, B. Orthen, B. Honnert, E. Jankovska et al., Workplace exposures to nanoparticles. Brussels, Belgium: European Agency for Safety and Health at Work (EU-OSHA), 2009.

, Communication from the Commission to the European Parliament, the Council and the European Economic and Social Committee, 2012.

R. Barouki, P. D. Gluckman, P. Grandjean, M. Hanson, and J. J. Heindel, Developmental origins of non-communicable disease: implications for research and public health, Environ Health, vol.11, p.42, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00712586

K. S. Hougaard and L. Campagnolo, Reproductive toxicity of engineered nanoparticles, pp.225-273, 2012.

C. M. Powers, A. S. Bale, A. D. Kraft, S. L. Makris, J. Trecki et al., Developmental neurotoxicity of engineered nanomaterials: identifying research needs to support human health risk assessment, Toxicol Sci, vol.134, issue.2, pp.225-267, 2013.

C. L. Klein, K. Wiench, M. Wiemann, L. Ma-hock, R. B. Van et al., Hazard identification of inhaled nanomaterials: making use of short-term inhalation studies, Arch Toxicol, vol.86, issue.7, pp.1137-51, 2012.

A. Pietroiusti, L. Campagnolo, and B. Fadeel, Interactions of engineered nanoparticles with organs protected by internal biological barriers, Small, vol.9, issue.9, pp.1557-72, 2013.

K. S. Hougaard, B. Fadeel, M. Gulumian, V. E. Kagan, and K. Savolainen, Developmental toxicity of engineered nanoparticles, pp.269-90, 2011.

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, Part Fibre Toxicol, vol.7, p.16, 2010.

P. Jackson, K. S. Hougaard, A. M. Boisen, N. R. Jacobsen, K. A. Jensen et al., Pulmonary exposure to carbon black by inhalation or instillation in pregnant mice: effects on liver DNA strand breaks in dams and offspring, Nanotoxicology, vol.6, issue.5, pp.484-500, 2011.

P. Moller, N. R. Jacobsen, J. K. Folkmann, P. H. Danielsen, L. Mikkelsen et al., Role of oxidative damage in toxicity of particulates, Free Radic Res, vol.44, issue.1, pp.1-46, 2010.

P. G. Wells, Y. Bhuller, C. S. Chen, W. Jeng, S. Kasapinovic et al., Molecular and biochemical mechanisms in teratogenesis involving reactive oxygen species, Toxicol Appl Pharmacol, vol.207, issue.2, pp.354-66, 2005.

U. Ratnayake, T. Quinn, D. W. Walker, and H. Dickinson, Cytokines and the neurodevelopmental basis of mental illness, Front Neurosci, vol.7, p.180, 2013.

F. J. Karsch, D. F. Battaglia, K. M. Breen, N. Debus, and T. G. Harris, Mechanisms for ovarian cycle disruption by immune/inflammatory stress, Stress, vol.5, issue.2, pp.101-113, 2002.

I. Iavicoli, L. Fontana, V. Leso, and A. Bergamaschi, The effects of nanomaterials as endocrine disruptors, Int J Mol Sci, vol.14, issue.8, pp.16732-801, 2013.

J. Sun, Q. Zhang, Z. Wang, and B. Yan, Effects of nanotoxicity on female reproductivity and fetal development in animal models, Int J Mol Sci, vol.14, issue.5, pp.9319-9356, 2013.

P. S. Shah and T. Balkhair, Air pollution and birth outcomes: a systematic review, Environ Int, vol.37, issue.2, pp.498-516, 2011.

S. Kannan, D. P. Misra, J. T. Dvonch, and A. Krishnakumar, Exposures to airborne particulate matter and adverse perinatal outcomes: a biologically plausible mechanistic framework for exploring potential, Cien Saude Colet, vol.12, issue.6, pp.1591-602, 2007.

K. S. Hougaard, K. A. Jensen, P. Nordly, C. Taxvig, U. Vogel et al., Effects of prenatal exposure to diesel exhaust particles on postnatal development, behavior, genotoxicity and inflammation in mice, Part Fibre Toxicol, vol.5, p.3, 2008.

M. Ema, M. Naya, M. Horimoto, and H. Kato, Developmental toxicity of diesel exhaust: a review of studies in experimental animals, Reprod Toxicol, vol.42, pp.1-17, 2013.

, Commission recommendation of 18 October on the definition of nanomaterial, vol.275, pp.38-40, 2011.

, European Commission Enterprise and Industry. Nanomaterials. European Commission. 2-2-2015

W. Prutner, Hazard and risk assessment of teratogenic chemicals under REACH, Methods Mol Biol, vol.947, pp.517-560, 2013.

S. Hankin, S. Peters, C. A. Poland, F. Hansen, S. Holmquist et al., Specific advice on fulfilling information requirements for nanomaterials under REACH (RIP-oN 2). Final project report

, Guidance on information requirements and chemical safety assessment. Appendix R7-1. Recommendations for nanomaterials applicable to chapter R7a Endpoint specific guidance, 2012.

. Oecd and . Oecd, Combined repeated dose toxicity study with the reproduction/developmental toxicity screening test, vol.422, 1996.

C. C. Lawson, B. Grajewski, G. P. Daston, L. M. Frazier, D. Lynch et al., Workgroup report: Implementing a national occupational reproductive research agenda -decade one and beyond, Environ Health Perspect, vol.114, issue.3, pp.435-476, 2006.

, Department of Health and Human Services. Safety of nanomaterials in cosmetic products. Guidance for industry. U.S. Food and Drug Administration, Nanotechnology Fact Sheet. U.S. Food and Drug Administration, 2014.

, OECD's Working Party on Nanotechnology, 2015.

V. Stone, S. Pozzi-mucelli, L. Tran, K. Aschberger, S. Sabella et al., ITS-NANO -prioritising nanosafety research to develop a stakeholder driven intelligent testing strategy, Part Fibre Toxicol, vol.11, p.9, 2014.

J. H. Arts, M. Hadi, A. M. Keene, R. Kreiling, D. Lyon et al., A critical appraisal of existing concepts for the grouping of nanomaterials, Regul Toxicol Pharmacol, vol.70, issue.2, pp.492-506, 2014.

K. Van-der-jagt, S. Munn, J. Torslov, and J. De-bruijn, Alternative approaches can reduce the use of test animals under REACH, European Commission Joint Research Centre, 2004.

F. R. Cassee, H. Muijser, E. Duistermaat, J. J. Freijer, K. B. Geerse et al., Particle size-dependent total mass deposition in lungs determines inhalation toxicity of cadmium chloride aerosols in rats. Application of a multiple path dosimetry model, Arch Toxicol, vol.76, issue.5-6, pp.277-86, 2002.

, Human respiratory tract model for radiological protection. A report of a Task Group of the International Commission of Radiological Protection, 66. ICRP publication, ICRP, 1994.

W. G. Kreyling, S. Hirn, W. Moller, C. Schleh, A. Wenk et al., Air-blood barrier translocation of tracheally instilled gold nanoparticles inversely depends on particle size, ACS Nano, vol.8, issue.1, pp.222-255, 2014.

B. Gulson, M. Mccall, M. Korsch, L. Gomez, P. Casey et al., Small amounts of zinc from zinc oxide particles in sunscreens applied outdoors are absorbed through human skin, Toxicol Sci, vol.118, issue.1, pp.140-149, 2010.

P. Krystek, J. Tentschert, Y. Nia, B. Trouiller, L. Noel et al., Method development and inter-laboratory comparison about the determination of titanium from titanium dioxide nanoparticles in tissues by inductively coupled plasma mass spectrometry, Anal Bioanal Chem, vol.406, issue.16, pp.3853-61, 2014.
URL : https://hal.archives-ouvertes.fr/ineris-01855499

W. H. De-jong, W. I. Hagens, P. Krystek, M. C. Burger, A. J. Sips et al., Particle size-dependent organ distribution of gold nanoparticles after intravenous administration, Biomaterials, vol.29, issue.12, pp.1912-1921, 2008.

D. P. Lankveld, A. G. Oomen, P. Krystek, A. Neigh, J. A. Troost-de et al., The kinetics of the tissue distribution of silver nanoparticles of different sizes, Biomaterials, vol.31, issue.32, pp.8350-61, 2010.

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, Part Fibre Toxicol, vol.11, p.30, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01143801

S. Hirn, M. Semmler-behnke, C. Schleh, A. Wenk, J. Lipka et al., Particle size-dependent and surface charge-dependent biodistribution of gold nanoparticles after intravenous administration, Eur J Pharm Biopharm, vol.77, issue.3, pp.407-423, 2011.

M. Semmler-behnke, W. G. Kreyling, J. Lipka, S. Fertsch, A. Wenk et al., Biodistribution of 1.4-and 18-nm gold particles in rats, Small, vol.4, issue.12, pp.2108-2119, 2008.

T. Cedervall, I. Lynch, S. Lindman, T. Berggard, E. Thulin et al., Understanding the nanoparticle-protein corona using methods to quantify exchange rates and affinities of proteins for nanoparticles, Proc Natl Acad Sci, vol.104, issue.7, pp.2050-2055, 2007.

A. Lesniak, F. Fenaroli, M. P. Monopoli, C. Aberg, K. A. Dawson et al., Effects of the presence or absence of a protein corona on silica nanoparticle uptake and impact on cells, ACS Nano, vol.6, issue.7, pp.5845-57, 2012.

D. P. Lankveld, R. G. Rayavarapu, P. Krystek, A. G. Oomen, H. W. Verharen et al., Blood clearance and tissue distribution of PEGylated and non-PEGylated gold nanorods after intravenous administration in rats, Nanomedicine (London), vol.6, issue.2, pp.339-388, 2011.

H. Yang, C. Sun, Z. Fan, X. Tian, L. Yan et al., Effects of gestational age and surface modification on materno-fetal transfer of nanoparticles in murine pregnancy, Sci Rep, vol.2, p.847, 2012.

P. Chavatte-palmer and M. Guillomot, Comparative implantation and placentation, Gynecol Obstet Invest, vol.64, issue.3, pp.166-74, 2007.
URL : https://hal.archives-ouvertes.fr/hal-02653359

F. Wooding and G. Burton, Implantation, maternofetal exchange and vascular relationships, pp.47-80, 2008.

A. M. Carter, Evolution of placental function in mammals: the molecular basis of gas and nutrient transfer, hormone secretion, and immune responses, Physiol Rev, vol.92, issue.4, pp.1543-76, 2012.

J. Guibourdenche, T. Fournier, and A. Malassine, Evain-Brion D. Development and hormonal functions of the human placenta, Folia Histochem Cytobiol, vol.47, issue.5, pp.35-40, 2009.

A. Tarrade, P. Chavatte-palmer, M. Guillomot, S. Camous, D. L. Evian-brion et al., Reproduction chez l'homme et l'animal. INRA, Chastant-Maillard S, pp.367-94, 2014.

A. Malassine and J. L. Frendo, Evain-Brion D. A comparison of placental development and endocrine functions between the human and mouse model, Hum Reprod Update, vol.9, issue.6, pp.531-540, 2003.

A. M. Carter, A. C. Enders, C. J. Jones, A. Mess, C. Pfarrer et al., Comparative placentation and animal models: patterns of trophoblast invasiona workshop report, Placenta, vol.27, pp.30-33, 2006.

M. Rubinchik-stern and S. Eyal, Drug interactions at the human placenta: what is the evidence?, Front Pharmacol, vol.3, p.126, 2012.

L. Cartwright, M. S. Poulsen, H. M. Nielsen, G. Pojana, L. E. Knudsen et al., In vitro placental model optimization for nanoparticle transport studies, Int J Nanomed, vol.7, pp.497-510, 2012.

H. Ali, I. Kalashnikova, M. A. White, M. Sherman, and E. Rytting, Preparation, characterization, and transport of dexamethasone-loaded polymeric nanoparticles across a human placental in vitro model, Int J Pharm, vol.454, issue.1, pp.149-57, 2013.

M. S. Poulsen, T. Mose, M. L. Leth, L. Mathiesen, K. L. Ehlert et al., Kinetics of silica nanoparticles in the human placenta, Nanotoxicology, vol.9, pp.79-86, 2015.

P. Wick, A. Malek, P. Manser, D. Meili, X. Maeder-althaus et al., Barrier capacity of human placenta for nanosized materials, Environ Health Perspect, vol.118, issue.3, pp.432-438, 2010.

S. C. Sumner, T. R. Fennell, R. W. Snyder, G. F. Taylor, and A. H. Lewin, Distribution of carbon-14 labeled C60 ([ 14 C]C60) in the pregnant and in the lactating dam and the effect of C60 exposure on the biochemical profile of urine, J Appl Toxicol, vol.30, issue.4, pp.354-60, 2010.

W. Qi, J. Bi, X. Zhang, J. Wang, J. Wang et al., Damaging effects of multiwalled carbon nanotubes on pregnant mice with different pregnancy times, Sci Rep, vol.4, p.4352, 2014.

M. Chu, Q. Wu, H. Yang, R. Yuan, S. Hou et al., Transfer of quantum dots from pregnant mice to pups across the placental barrier, Small, vol.6, issue.5, pp.670-678, 2010.

J. L. Blum, J. Q. Xiong, C. Hoffman, and J. T. Zelikoff, Cadmium associated with inhaled cadmium oxide nanoparticles impacts fetal and neonatal development and growth, Toxicol Sci, vol.26, issue.2, pp.478-86, 2012.

K. Yamashita, Y. Yoshioka, K. Higashisaka, K. Mimura, Y. Morishita et al., Silica and titanium dioxide nanoparticles cause pregnancy complications in mice, Nat Nanotechnol, vol.6, issue.5, pp.321-329, 2011.

K. Rattanapinyopituk, A. Shimada, T. Morita, M. Sakurai, A. Asano et al., Demonstration of the clathrin-and caveolin-mediated endocytosis at the maternal-fetal barrier in mouse placenta after intravenous administration of gold nanoparticles, J Vet Med Sci, vol.76, issue.3, pp.377-87, 2014.

D. Bona, K. R. Xu, Y. Ramirez, P. A. , D. J. Parker et al., Surface charge and dosage dependent potential developmental toxicity and biodistribution of iron oxide nanoparticles in pregnant CD-1 mice, Reprod Toxicol, vol.50, pp.36-42, 2014.

S. Grafmuller, P. Manser, H. F. Krug, P. Wick, and V. Mu, Determination of the transport rate of xenobiotics and nanomaterials across the placenta using the ex vivo human placental perfusion model, J Vis Exp, vol.76, p.50401, 2013.

C. S. Correia, L. Walker, K. Paul, and M. Saunders, The toxicity, transport and uptake of nanoparticles in the in vitro BeWo b30 placental cell barrier model used within NanoTEST, Nanotoxicology, vol.9, pp.66-78, 2013.

L. Mathiesen, T. A. Morck, G. Zuri, M. H. Andersen, C. Pehrson et al., Modelling of human transplacental transport as performed in, Basic Clin Pharmacol Toxicol, vol.115, pp.93-100, 2014.

X. Tian, M. Zhu, L. Du, J. Wang, Z. Fan et al., Intrauterine inflammation increases materno-fetal transfer of gold nanoparticles in a size-dependent manner in murine pregnancy, Small, vol.9, issue.14, pp.2432-2441, 2013.

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.121-154, 2012.

A. Erdely, A. Liston, R. Salmen-muniz, T. Hulderman, S. H. Young et al., Identification of systemic markers from a pulmonary carbon nanotube exposure, J Occup Environ Med, vol.53, issue.6, pp.80-86, 2011.

A. T. Saber, J. S. Lamson, N. R. Jacobsen, G. Ravn-haren, K. S. Hougaard et al., Particle-induced pulmonary acute phase response correlates with neutrophil influx linking inhaled particles and cardiovascular risk, PLOS ONE, vol.8, issue.7, p.69020, 2013.

K. Koga, G. Izumi, G. Mor, T. Fujii, and Y. Osuga, Toll-like receptors at the maternal-fetal interface in normal pregnancy and pregnancy complications, Am J Reprod Immunol, vol.72, issue.2, pp.192-205, 2014.

C. Zhao, J. Liao, W. Chu, S. Wang, T. Yang et al., Involvement of TLR2 and TLR4 and Th1/Th2 shift in inflammatory responses induced by fine ambient particulate matter in mice, Inhal Toxicol, vol.24, issue.13, pp.918-945, 2012.

A. Pietroiusti, M. Massimiani, I. Fenoglio, M. Colonna, F. Valentini et al., Low doses of pristine and oxidized single-wall carbon nanotubes affect Mammalian embryonic development, ACS Nano, vol.5, issue.6, pp.4624-4657, 2011.

D. J. Barker, In utero programming of chronic disease, Clin Sci (Colch), vol.95, issue.2, pp.115-143, 1998.

R. L. Jirtle and M. K. Skinner, Environmental epigenomics and disease susceptibility, Nat Rev Genet, vol.8, issue.4, pp.253-62, 2007.

M. Collotta, P. A. Bertazzi, and V. Bollati, Epigenetics and pesticides, Toxicology, vol.307, pp.35-41, 2013.

D. J. Barker, The developmental origins of adult disease, J Am Coll Nutr, vol.23, issue.6, pp.588-95, 2004.

N. Khansari, Y. Shakiba, and M. Mahmoudi, Chronic inflammation and oxidative stress as a major cause of age-related diseases and cancer, Recent Pat Inflamm Allergy Drug Discov, vol.3, issue.1, pp.73-80, 2009.

P. A. Dennery, Oxidative stress in development: nature or nurture? Free Radic, Biol Med, vol.49, issue.7, pp.1147-51, 2010.

J. M. Hansen and C. Harris, Redox control of teratogenesis, Reprod Toxicol, vol.35, pp.165-79, 2013.

N. A. Hodyl, M. J. Stark, A. Osei-kumah, and V. L. Clifton, Prenatal programming of the innate immune response following in utero exposure to inflammation: a sexually dimorphic process?, Expert Rev Clin Immunol, vol.7, issue.5, pp.579-92, 2011.

K. Donaldson, D. Brown, A. Clouter, R. Duffin, W. Macnee et al., The pulmonary toxicology of ultrafine particles, J Aerosol Med, vol.15, issue.2, pp.213-233, 2002.

K. Shirasuna, F. Usui, T. Karasawa, H. Kimura, A. Kawashima et al., Nanosilica-induced placental inflammation and pregnancy complications: different roles of the inflammasome components NLRP3 and ASC, Nanotoxicology, vol.11, pp.1-14, 2014.

T. Fujitani, K. Ohyama, A. Hirose, T. Nishimura, D. Nakae et al., Teratogenicity of multi-wall carbon nanotube (MWCNT) in ICR mice, J Toxicol Sci, vol.37, issue.1, pp.81-90, 2012.

K. J. Lim, O. A. Odukoya, T. C. Li, and I. D. Cooke, Cytokines and immuno-endocrine factors in recurrent miscarriage, Hum Reprod Update, vol.2, issue.6, pp.469-81, 1996.

J. O. De-melo, S. F. Soto, I. A. Katayama, C. F. Wenceslau, A. G. Pires et al., Inhalation of fine particulate matter during pregnancy increased IL-4 cytokine levels in the fetal portion of the placenta, Toxicol Lett, vol.232, issue.2, pp.475-80, 2015.

A. Sood, S. Salih, D. Roh, L. Lacharme-lora, M. Parry et al., Signalling of DNA damage and cytokines across cell barriers exposed to nanoparticles depends on barrier thickness, Nat Nanotechnol, vol.6, issue.12, pp.824-857, 2011.

M. S. Hsieh, N. H. Shiao, and W. H. Chan, Cytotoxic effects of CdSe quantum dots on maturation of mouse oocytes, fertilization, and fetal development, Int J Mol Sci, vol.10, issue.5, pp.2122-2157, 2009.

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, Environ Mol Mutagen, vol.52, issue.6, pp.425-464, 2011.

P. Jackson, S. Halappanavar, K. S. Hougaard, A. Williams, A. M. Madsen et al., Maternal inhalation of surface-coated nanosized titanium dioxide (UV-Titan) in C57BL/6 mice: effects in prenatally exposed offspring on hepatic DNA damage and gene expression, Nanotoxicology, vol.7, pp.85-96, 2013.

P. Jackson, U. Vogel, H. Wallin, and K. S. Hougaard, Prenatal exposure to carbon black (Printex 90): effects on sexual development and neurofunction, Basic Clin Pharmacol Toxicol, vol.109, issue.6, pp.434-441, 2011.

P. Jackson, K. S. Hougaard, U. Vogel, D. Wu, L. Casavant et al., Exposure of pregnant mice to carbon black by intratracheal instillation: toxicogenomic effects in dams and offspring, Mutat Res, vol.745, issue.1-2, pp.73-83, 2012.

K. S. Hougaard, P. Jackson, Z. O. Kyjovska, R. K. Birkedal, P. J. De-temmerman et al., Effects of lung exposure to carbon nanotubes on female fertility and pregnancy. A study in mice, Reprod Toxicol, vol.41, pp.86-97, 2013.

N. Ono, S. Oshio, Y. Niwata, S. Yoshida, N. Tsukue et al., Prenatal exposure to diesel exhaust impairs mouse spermatogenesis, Inhal Toxicol, vol.19, issue.3, pp.275-81, 2007.

M. Umezawa, C. Sakata, N. Tanaka, M. Tabata, K. Takeda et al., Pathological study for the effects of in utero and postnatal exposure to diesel exhaust on a rat endometriosis model, J Toxicol Sci, vol.36, issue.4, pp.493-501, 2011.

C. S. Weldy, Y. Liu, H. D. Liggitt, and M. T. Chin, In utero exposure to diesel exhaust air pollution promotes adverse intrauterine conditions, resulting in weight gain, altered blood pressure, and increased susceptibility to heart failure in adult mice, PLOS ONE, vol.9, issue.2, p.88582, 2014.

A. Fujimoto, N. Tsukue, M. Watanabe, I. Sugawara, R. Yanagisawa et al., Diesel exhaust affects immunological action in the placentas of mice, Environ Toxicol, vol.20, issue.4, pp.431-471, 2005.

N. Watanabe and M. Kurita, The masculinization of the fetus during pregnancy due to inhalation of diesel exhaust, Environ Health Perspect, vol.109, issue.2, pp.111-120, 2001.

C. Li, X. Li, A. K. Suzuki, Y. Zhang, Y. Fujitani et al., Effects of exposure to nanoparticle-rich diesel exhaust on pregnancy in rats, J Reprod Dev, vol.59, issue.2, pp.145-50, 2013.

N. Watanabe, Decreased number of sperms and Sertoli cells in mature rats exposed to diesel exhaust as fetuses, Toxicol Lett, vol.155, issue.1, pp.51-59, 2005.

N. Watanabe and M. Ohsawa, Elevated serum immunoglobulin E to Cryptomeria japonica pollen in rats exposed to diesel exhaust during fetal and neonatal periods, BMC Pregnancy Childbirth, vol.2, issue.1, pp.2-11, 2002.

W. E. Pepelko and W. B. Peirano, Health effects of exposure to diesel engine emissions: a summary of animal studies conducted by the US Environmental Protection Agency's Health Effects Research Laboratories at Cincinnati, Ohio, J Am Coll Toxicol, vol.2, issue.4, pp.253-306, 1983.

N. Tsukue, H. Tsubone, and A. K. Suzuki, Diesel exhaust affects the abnormal delivery in pregnant mice and the growth of their young, Inhal Toxicol, vol.14, issue.6, pp.635-51, 2002.

, International Programme for Chemical Safety. Global Assessment of the Stateof-the-Science for Endocrine Disruptors. Geneva: World Health Organisation, 2002.

N. Tsukue, M. Watanabe, T. Kumamoto, H. Takano, and K. Takeda, Perinatal exposure to diesel exhaust affects gene expression in mouse cerebrum, Arch Toxicol, vol.83, issue.11, pp.985-1000, 2009.

C. Li, S. Taneda, K. Taya, G. Watanabe, X. Li et al., Effects of in utero exposure to nanoparticle-rich diesel exhaust on testicular function in immature male rats, Toxicol Lett, vol.185, issue.1, pp.1-8, 2009.

J. L. Tang-peronard, H. R. Andersen, T. K. Jensen, and B. L. Heitmann, Endocrinedisrupting chemicals and obesity development in humans: a review, Obes Rev, vol.12, issue.8, pp.622-658, 2011.

A. Janesick and B. Blumberg, Obesogens, stem cells and the developmental programming of obesity, Int J Androl, vol.35, issue.3, pp.437-485, 2012.

E. C. Cottrell and S. E. Ozanne, Developmental programming of energy balance and the metabolic syndrome, Proc Nutr Soc, vol.66, issue.2, pp.198-206, 2007.

A. Onoda, M. Umezawa, K. Takeda, T. Ihara, and M. Sugamata, Effects of maternal exposure to ultrafine carbon black on brain perivascular macrophages and surrounding astrocytes in offspring mice, PLOS ONE, vol.9, issue.4, p.94336, 2014.

M. Sugamata, T. Ihara, H. Takano, S. Oshio, and K. Takeda, Maternal diesel exhaust exposure damages newborn murine brains, J Health Sci, vol.52, issue.1, pp.82-86, 2006.

M. Sugamata, T. Ihara, M. Sugamata, and K. Takeda, Maternal exposure to diesel exhaust leads to pathological similarity of autism in newborns, J Health Sci, vol.52, issue.4, pp.486-494, 2006.

S. Yokota, K. Mizuo, N. Moriya, S. Oshio, I. Sugawara et al., Effect of prenatal exposure to diesel exhaust on dopaminergic system in mice, Neurosci Lett, vol.449, issue.1, pp.38-41, 2009.

T. Suzuki, S. Oshio, M. Iwata, H. Saburi, T. Odagiri et al., In utero exposure to a low concentration of diesel exhaust affects spontaneous locomotor activity and monoaminergic system in male mice, Part Fibre Toxicol, vol.7, p.7, 2010.

S. Yokota, N. Moriya, M. Iwata, M. Umezawa, S. Oshio et al., Exposure to diesel exhaust during fetal period affects behavior and neurotransmitters in male offspring mice, J Toxicol Sci, vol.38, issue.1, pp.13-23, 2013.

R. K. Thirtamara, S. Doherty-lyons, C. Bolden, D. Willis, C. Hoffman et al., Prenatal and early-life exposure to high-level diesel exhaust particles leads to increased locomotor activity and repetitive behaviors in mice, Autism Res, vol.6, issue.4, pp.248-57, 2013.

J. L. Bolton, S. H. Smith, N. C. Huff, M. I. Gilmour, W. M. Foster et al., Prenatal air pollution exposure induces neuroinflammation and predisposes offspring to weight gain in adulthood in a sex-specific manner, FASEB J, vol.26, issue.11, pp.4743-54, 2012.

D. A. Davis, M. Bortolato, S. C. Godar, T. K. Sander, N. Iwata et al., Prenatal exposure to urban air nanoparticles in mice causes altered neuronal differentiation and depression-like responses, PLOS ONE, vol.8, issue.5, p.64128, 2013.

S. Yoshida, K. Hiyoshi, S. Oshio, H. Takano, K. Takeda et al., Effects of fetal exposure to carbon nanoparticles on reproductive function in male offspring, Fertil Steril, vol.93, issue.5, pp.1695-1704, 2009.

Z. O. Kyjovska, A. M. Boisen, P. Jackson, H. Wallin, U. Vogel et al., Daily sperm production: application in studies of prenatal exposure to nanoparticles in mice, Reprod Toxicol, vol.36, pp.88-97, 2013.

K. Takeda, K. 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, issue.1, pp.95-102, 2009.

J. Bowles and P. Koopman, Retinoic acid, meiosis and germ cell fate in mammals, Development, vol.134, pp.3401-3412, 2007.

A. J. Childs, G. Cowan, H. L. Kinnell, R. A. Anderson, and P. T. Saunders, Retinoic acid signalling and the control of meiotic entry in the human fetal gonad, PLoS ONE, vol.6, issue.6, p.20249, 2011.

J. E. Seo, G. Kwon, J. T. Lee, M. Lee, B. Eom et al., Exposure to zinc oxide nanoparticles affects reproductive development and biodistribution in offspring rats, J Toxicol Sci, vol.38, issue.4, pp.525-555, 2013.

J. G. Hemmingsen, K. S. Hougaard, C. Talsness, A. Wellejus, S. Loft et al., Prenatal exposure to diesel exhaust particles and effect on the male reproductive system in mice, Toxicology, vol.264, issue.1-2, pp.61-69, 2009.

Y. S. El-sayed, R. Shimizu, A. Onoda, K. Takeda, and M. Umezawa, Carbon black nanoparticle exposure during middle and late fetal development induces immune activation in male offspring mice, Toxicology, vol.327, pp.53-61, 2015.

J. S. Hansen, H. Johansson, U. Vogel, S. T. Larsen, and K. S. Hougaard, Maternal exposure to carbon nanotubes and effects on allergy in offspring. In: 7th meeting of the immunotoxicology and chemical allergy section (ITCASS), 2012.

A. V. Fedulov, A. Leme, Z. Yang, M. Dahl, R. Lim et al., Pulmonary exposure to particles during pregnancy causes increased neonatal asthma susceptibility, Am J Respir Cell Mol Biol, vol.38, issue.1, pp.57-67, 2008.

Y. Zhang, L. Mikhaylova, L. Kobzik, and A. V. Fedulov, Estrogen-mediated impairment of macrophageal uptake of environmental TiO particles to explain inflammatory effect of TiO on airways during pregnancy, J Immunotoxicol, vol.12, issue.1, pp.81-91, 2015.

S. Manners, R. Alam, D. A. Schwartz, and M. M. Gorska, A mouse model links asthma susceptibility to prenatal exposure to diesel exhaust, J Allergy Clin Immunol, vol.134, issue.1, pp.63-72, 2014.

L. Corson, H. Zhu, C. Quan, G. Grunig, M. Ballaney et al., Prenatal allergen and diesel exhaust exposure and their effects on allergy in adult offspring mice, Allergy Asthma Clin Immunol, vol.6, issue.1, p.7, 2010.

T. Sharkhuu, D. L. Doerfler, Q. T. Krantz, R. W. Luebke, W. P. Linak et al., Effects of prenatal diesel exhaust inhalation on pulmonary inflammation and development of specific immune responses, Toxicol Lett, vol.196, issue.1, pp.12-20, 2010.

X. Hong, C. Liu, X. Chen, Y. Song, Q. Wang et al., Maternal exposure to airborne particulate matter causes postnatal immunological dysfunction in mice offspring, Toxicology, vol.306, pp.59-67, 2013.

R. L. Auten, E. N. Potts, S. N. Mason, B. Fischer, Y. Huang et al., Maternal exposure to particulate matter increases postnatal ozone-induced airway hyperreactivity in juvenile mice, Am J Respir Crit Care Med, vol.180, issue.12, pp.1218-1244, 2009.

R. L. Auten, M. I. Gilmour, Q. T. Krantz, E. N. Potts, S. N. Mason et al., Maternal diesel inhalation increases airway hyperreactivity in ozone-exposed offspring, Am J Respir Cell Mol Biol, vol.46, issue.4, pp.454-60, 2012.

J. S. Hansen, T. Alberg, H. Rasmussen, M. Lovik, and U. C. Nygaard, Determinants of experimental allergic responses: interactions between allergen dose, sex and age, Scand J Immunol, vol.10, p.3083, 2011.

D. Zhou and Y. X. Pan, Pathophysiological basis for compromised health beyond generations: role of maternal high-fat diet and low-grade chronic inflammation, J Nutr Biochem, vol.26, issue.1, pp.1-8, 2015.

P. A. Stapleton, V. C. Minarchick, J. Yi, K. Engels, C. R. Mcbride et al., Maternal engineered nanomaterial exposure and fetal microvascular function: does the Barker hypothesis apply?, Am J Obstet Gynecol, vol.209, issue.3, pp.227-311, 2013.

P. A. Stapleton, C. E. Nichols, J. Yi, C. R. Mcbride, V. C. Minarchick et al., Microvascular and mitochondrial dysfunction in the female F1 generation after gestational TiO nanoparticle exposure, Nanotoxicology, vol.9, p.5, 2012.

M. Umezawa, S. Kudo, S. Yanagita, Y. Shinkai, R. Niki et al., Maternal exposure to carbon black nanoparticle increases collagen type VIII expression in the kidney of offspring, J Toxicol Sci, vol.36, issue.4, pp.461-469, 2011.

N. R. Damaceno-rodrigues, M. M. Veras, E. M. Negri, A. C. Zanchi, C. R. Rhoden et al., Effect of pre-and postnatal exposure to urban air pollution on myocardial lipid peroxidation levels in adult mice, Inhal Toxicol, vol.21, issue.13, pp.1129-1166, 2009.

C. S. Weldy, Y. Liu, Y. C. Chang, I. O. Medvedev, J. R. Fox et al., In utero and early life exposure to diesel exhaust air pollution increases adult susceptibility to heart failure in mice, Part Fibre Toxicol, vol.10, issue.1, p.59, 2013.

M. W. Gorr, M. Velten, T. D. Nelin, D. J. Youtz, Q. Sun et al., Early life exposure to air pollution induces adult cardiac dysfunction, Am J Physiol Heart Circ Physiol, vol.307, issue.9, pp.1353-60, 2014.

J. A. Bourdon, A. T. Saber, N. R. Jacobsen, K. A. Jensen, A. M. Madsen et al., Carbon black nanoparticle instillation induces sustained inflammation and genotoxicity in mouse lung and liver, Part Fibre Toxicol, vol.9, issue.1, p.5, 2012.

K. I. Aston and D. T. Carrell, Emerging evidence for the role of genomic instability in male factor infertility, Syst Biol Reprod Med, vol.58, issue.2, pp.71-80, 2012.

A. M. Boisen, T. Shipley, P. Jackson, K. S. Hougaard, H. Wallin et al., UV-Titan) does not induce ESTR mutations in the germline of prenatally exposed female mice, Part Fibre Toxicol, vol.9, p.19, 2012.

I. D. Adler, A. Carere, U. Eichenlaub-ritter, and F. Pacchierotti, Gender differences in the induction of chromosomal aberrations and gene mutations in rodent germ cells, Environ Res, vol.104, issue.1, pp.37-45, 2007.

A. M. Boisen, T. Shipley, P. Jackson, H. Wallin, C. Nellemann et al., In utero exposure to nanosized carbon black (Printex90) does not induce tandem repeat mutations in female murine germ cells, Reprod Toxicol, vol.41, pp.45-53, 2013.

C. Ritz, W. Ruminski, K. S. Hougaard, H. Wallin, U. Vogel et al., Germline mutation rates in mice following in utero exposure to diesel exhaust particles by maternal inhalation, Mutat Res, vol.712, issue.1-2, pp.55-63, 2011.

C. Yauk, A. Polyzos, A. Rowan-carroll, C. M. Somers, R. W. Godschalk et al., Germ-line mutations, DNA damage, and global hypermethylation in mice exposed to particulate air pollution in an urban/industrial location, Proc Natl Acad Sci U S A, vol.105, issue.2, pp.605-615, 2008.

C. M. Somers, C. Yauk, P. A. White, C. L. Parfett, and J. S. Quinn, Air pollution induces heritable DNA mutations, Proc Natl Acad Sci U S A, vol.99, issue.25, pp.15904-15911, 2002.

C. A. Austin, T. H. Umbreit, K. M. Brown, D. S. Barber, B. J. Dair et al., Distribution of silver nanoparticles in pregnant mice and developing embryos, Nanotoxicology, vol.6, pp.912-934, 2012.

R. Maeda-mamiya, E. Noiri, H. Isobe, W. Nakanishi, K. Okamoto et al., In vivo gene delivery by cationic tetraamino fullerene, Proc Natl Acad Sci, vol.107, issue.12, pp.5339-5383, 2010.

L. Campagnolo, M. Massimiani, G. Palmieri, R. Bernardini, C. Sacchetti et al., Biodistribution and toxicity of pegylated single wall carbon nanotubes in pregnant mice, Part Fibre Toxicol, vol.10, issue.1, p.21, 2013.

X. Huang, F. Zhang, X. Sun, K. Y. Choi, G. Niu et al., The genotypedependent influence of functionalized multiwalled carbon nanotubes on fetal development, Biomaterials, vol.35, issue.2, pp.856-65, 2014.

N. A. Philbrook, L. M. Winn, A. R. Afrooz, N. B. Saleh, and V. K. Walker, The effect of TiO2 and Ag nanoparticles on reproduction and development of Drosophila melanogaster and CD-1 mice, Toxicol Appl Pharmacol, vol.257, issue.3, pp.429-465, 2011.

S. K. Mwilu, E. Badawy, A. M. Bradham, K. Nelson, C. Thomas et al., Changes in silver nanoparticles exposed to human synthetic stomach fluid: effects of particle size and surface chemistry, Sci Total Environ, vol.447, pp.90-98, 2013.

N. A. Philbrook, V. K. Walker, A. R. Afrooz, N. B. Saleh, and L. M. Winn, Investigating the effects of functionalized carbon nanotubes on reproduction and development in Drosophila melanogaster and CD-1 mice, Reprod Toxicol, vol.32, issue.4, pp.442-450, 2011.

J. S. Hong, S. Kim, S. H. Lee, J. E. Lee, B. Yoon et al., Combined repeated-dose toxicity study of silver nanoparticles with the reproduction/developmental toxicity screening test, Nanotoxicology, vol.8, issue.4, pp.349-62, 2014.

E. Sadauskas, H. Wallin, M. Stoltenberg, U. Vogel, P. Doering et al., Kupffer cells are central in the removal of nanoparticles from the organism, Part Fibre Toxicol, vol.4, p.10, 2007.

T. Tsuchiya, I. Oguri, Y. N. Yamakoshi, and N. Miyata, Novel harmful effects of [60]fullerene on mouse embryos in vitro and in vivo, FEBS Lett, vol.393, issue.1, pp.139-184, 1996.

U. Gehring, L. Tamburic, H. Sbihi, H. W. Davies, and M. Brauer, Impact of noise and air pollution on pregnancy outcomes, Epidemiology, vol.25, issue.3, pp.351-359, 2014.

P. Dadvand, J. Parker, M. L. Bell, M. Bonzini, M. Brauer et al., Maternal exposure to particulate air pollution and term birth weight: a multi-country evaluation of effect and heterogeneity, Environ Health Perspect, vol.121, issue.3, pp.267-373, 2013.

M. Pedersen, L. Stayner, R. Slama, M. Sorensen, F. Figueras et al., Ambient air pollution and pregnancy-induced hypertensive disorders: a systematic review and meta-analysis, Hypertension, vol.64, issue.3, pp.494-500, 2014.

T. Yorifuji, H. Naruse, S. Kashima, T. Murakoshi, and H. Doi, Residential proximity to major roads and obstetrical complications, Sci Total Environ, vol.508, pp.188-92, 2015.

R. Slama, F. Ballester, M. Casas, S. Cordier, M. Eggesbo et al., Epidemiologic tools to study the influence of environmental factors on fecundity and pregnancy-related outcomes, Epidemiol Rev, vol.36, issue.1, pp.148-64, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01002724

M. Vrijheid, D. Martinez, S. Manzanares, P. Dadvand, A. Schembari et al., Ambient air pollution and risk of congenital anomalies: a systematic review and meta-analysis, Environ Health Perspect, vol.119, issue.5, pp.598-606, 2011.

E. K. Chen, D. Zmirou-navier, C. Padilla, and S. Deguen, Effects of air pollution on the risk of congenital anomalies: a systematic review and meta-analysis, Int J Environ Res Public Health, vol.11, issue.8, pp.7642-68, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01118609

A. Rahmalia, L. Giorgis-allemand, J. Lepeule, C. Philippat, J. Galineau et al., Pregnancy exposure to atmospheric pollutants and placental weight: an approach relying on a dispersion model, Environ Int, vol.48, pp.47-55, 2012.

E. H. Van-den-hooven, F. H. Pierik, K. Y. De, A. Hofman, S. W. Van-ratingen et al., Air pollution exposure and markers of placental growth and function: the generation R study, Environ Health Perspect, vol.120, issue.12, pp.1753-1762, 2012.

T. Yorifuji, H. Naruse, S. Kashima, T. Murakoshi, T. Tsuda et al., Residential proximity to major roads and placenta/birth weight ratio, Sci Total Environ, vol.414, pp.98-102, 2012.

N. Baiz, R. Slama, M. C. Bene, M. A. Charles, M. N. Kolopp-sarda et al., Maternal exposure to air pollution before and during pregnancy related to changes in newborn's cord blood lymphocyte subpopulations. The EDEN study cohort, BMC Pregnancy Childbirth, vol.11, p.87, 2011.

I. Hertz-picciotto, C. E. Herr, P. S. Yap, M. Dostal, R. H. Shumway et al., Air pollution and lymphocyte phenotype proportions in cord blood, Environ Health Perspect, vol.113, issue.10, pp.1391-1399, 2005.

M. Guxens, R. Garcia-esteban, L. Giorgis-allemand, J. Forns, C. Badaloni et al., Air pollution during pregnancy and childhood cognitive and psychomotor development: six European birth cohorts, Epidemiology, vol.25, issue.5, pp.636-683, 2014.

W. Jedrychowski, F. P. Perera, U. Maugeri, D. Mrozek-budzyn, E. Mroz et al., Early wheezing phenotypes and severity of respiratory illness in very early childhood: study on intrauterine exposure to fine particle matter, Environ Int, vol.35, issue.6, pp.877-84, 2009.

W. A. Jedrychowski, F. P. Perera, J. D. Spengler, E. Mroz, L. Stigter et al., Intrauterine exposure to fine particulate matter as a risk factor for increased susceptibility to acute broncho-pulmonary infections in early childhood, Int J Hyg Environ Health, vol.216, issue.4, pp.395-401, 2013.

E. Thiering, J. Cyrys, J. Kratzsch, C. Meisinger, B. Hoffmann et al., Long-term exposure to traffic-related air pollution and insulin resistance in children: results from the GINIplus and LISAplus birth cohorts, Diabetologia, vol.56, issue.8, pp.1696-704, 2013.

S. D. Adar, L. Sheppard, S. Vedal, J. F. Polak, P. D. Sampson et al., Fine particulate air pollution and the progression of carotid intima-medial thickness: a prospective cohort study from the multi-ethnic study of atherosclerosis and air pollution, PLoS Med, vol.10, issue.4, p.1001430, 2013.

C. H. Backes, T. Nelin, M. W. Gorr, and L. E. Wold, Early life exposure to air pollution: how bad is it?, Toxicol Lett, vol.216, issue.1, pp.47-53, 2013.

B. Fischer, P. Chavatte-palmer, C. Viebahn, S. A. Navarrete, and V. Duranthon, Rabbit as a reproductive model for human health, Reproduction, vol.144, issue.1, pp.1-10, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01019868

A. M. Carter, Animal models of human placentation -a review, Placenta, vol.28, pp.41-48, 2007.

G. Oberdörster, A. Maynard, K. Donaldson, V. Castranova, J. Fitzpatrick et al., Principles for characterizing the potential human health effects from exposure to nanomaterials: elements of a screening strategy, Part Fibre Toxicol, vol.2, p.8, 2005.

R. Landsiedel, L. Ma-hock, T. Hofmann, M. Wiemann, V. Strauss et al., Application of short-term inhalation studies to assess the inhalation toxicity of nanomaterials, Part Fibre Toxicol, vol.11, p.16, 2014.

K. Loeschner, N. Hadrup, K. Qvortrup, A. Larsen, X. Gao et al., Distribution of silver in rats following 28 days of repeated oral exposure to silver nanoparticles or silver acetate, Part Fibre Toxicol, vol.8, p.18, 2011.

, OECD Guideline for testing of chemicals 416. Two-generation reproduction toxicity study, 2001.

, OECD Guideline for testing of chemicals 443, 2012.

, OECD Guideline for testing of chemicals. 426. Developmental Neurotoxicity Study, 2007.

S. L. Makris, K. Raffaele, S. Allen, W. J. Bowers, U. Hass et al., A retrospective performance assessment of the developmental neurotoxicity study in support of OECD test guideline 426, Environ Health Perspect, vol.117, issue.1, pp.17-25, 2009.

, Nanotechnologies -compilation and description of toxicological screening methods for manufactured nanomaterials, 2014.

A. Tarrade, K. Schoonjans, L. Pavan, J. Auwerx, C. Rochette-egly et al., PPARgamma/RXRalpha heterodimers control human trophoblast invasion, J Clin Endocrinol Metab, vol.86, issue.10, pp.5017-5041, 2001.