M. Brandes, J. C. Verzijden, C. J. Hamilton, N. P. De-weys, J. P. De-bruin et al., Is the fertility treatment itself a risk factor for early pregnancy loss?, Reprod Biomed Online, vol.22, pp.192-201, 2011.

P. Chaveeva, I. F. Carbone, A. Syngelaki, R. Akolekar, and K. H. Nicolaides, Contribution of method of conception on pregnancy outcome after the 11-13 weeks scan, Fetal Diagn Ther, vol.30, pp.9-22, 2011.

J. X. Wang, R. J. Norman, and A. J. Wilcox, Incidence of spontaneous abortion among pregnancies produced by assisted reproductive technology, Hum Reprod, vol.19, pp.272-279, 2004.

L. Delle-piane, W. Lin, X. Liu, A. Donjacour, P. Minasi et al., Effect of the method of conception and embryo transfer procedure on mid-gestation placenta and fetal development in an Ivf mouse model, Hum Reprod, vol.25, pp.2039-2085, 2010.

P. Fauque, M. A. Ripoche, J. Tost, L. Journot, A. Gabory et al., Modulation of imprinted gene network in placenta results in normal development of in vitro manipulated mouse embryos, Hum Mol Genet, vol.19, pp.1779-90, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01788884

I. Van-der-auwera, D. 'hooghe, and T. , Superovulation of female mice delays embryonic and fetal development, Hum Reprod, vol.16, pp.1237-1280, 2001.

C. Thomopoulos, C. Tsioufis, H. Michalopoulou, T. Makris, V. Papademetriou et al., Assisted reproductive technology and pregnancy-related hypertensive complications: a systematic review, J Hum Hypertens, vol.27, pp.148-57, 2013.

R. A. Jackson, K. A. Gibson, Y. W. Wu, and M. S. Croughan, Perinatal outcomes in singletons following in vitro fertilization: a meta-analysis, Obstet Gynecol, vol.103, pp.551-63, 2004.

A. Pinborg, U. B. Wennerholm, L. B. Romundstad, A. Loft, K. Aittomaki et al., Why do singletons conceived after assisted reproduction technology have adverse perinatal outcome? Systematic review and meta-analysis, Hum Reprod Update, vol.19, pp.87-104, 2013.

E. Bloise, W. Lin, X. Liu, R. Simbulan, K. S. Kolahi et al., Impaired placental nutrient transport in mice generated by in vitro fertilization, Endocrinology, vol.153, pp.3457-67, 2012.

Z. Chen, K. M. Robbins, K. D. Wells, and R. M. Rivera, Large offspring syndrome: a bovine model for the human loss-of-imprinting overgrowth syndrome Beckwith-Wiedemann, Epigenetics, vol.8, pp.591-601, 2013.

A. T. Grazul-bilska, M. L. Johnson, P. P. Borowicz, L. Baranko, D. A. Redmer et al., Placental development during early pregnancy in sheep: effects of embryo origin on fetal and placental growth and global methylation, Theriogenology, vol.79, pp.94-102, 2013.

S. Hiendleder, C. Mund, H. D. Reichenbach, H. Wenigerkind, G. Brem et al., Tissue-specific elevated genomic cytosine methylation levels are associated with an overgrowth phenotype of bovine fetuses derived by in vitro techniques, Biol Reprod, vol.71, pp.217-240, 2004.

N. Hori, M. Nagai, M. Hirayama, T. Hirai, K. Matsuda et al., Aberrant Cpg methylation of the imprinting control region Kvdmr1 detected in assisted reproductive technology-produced calves and pathogenesis of large offspring syndrome, Anim Reprod Sci, vol.122, pp.303-315, 2010.

J. R. Miles, C. E. Farin, K. F. Rodriguez, J. E. Alexander, and P. W. Farin, Angiogenesis and morphometry of bovine placentas in late gestation from embryos produced in vivo or in vitro, Biol Reprod, vol.71, pp.1919-1945, 2004.

D. P-t-akge, . Go-s-t-in-oa-,t-o-s-ch-ip, A. Id-anza, F. Zacchini, and M. Czernik, Post-implantation mortality of in vitro produced embryos is associated with DNA methyltransferase 1 dysfunction in sheep placenta, Hum Reprod, vol.28, pp.298-305, 2013.

K. D. Sinclair, L. E. Young, I. Wilmut, and T. G. Mcevoy, In-utero overgrowth in ruminants following embryo culture: lessons from mice and a warning to men, Hum Reprod, vol.15, issue.5, pp.68-86, 2000.

L. E. Young, K. D. Sinclair, and I. Wilmut, Large offspring syndrome in cattle and sheep, Rev Reprod, vol.3, pp.155-63, 1998.

Y. Zhang, Y. L. Zhang, C. Feng, Y. T. Wu, A. X. Liu et al., Comparative proteomic analysis of human placenta derived from assisted reproductive technology, Proteomics, vol.8, pp.4344-56, 2008.

Q. Huang, J. Li, F. Wang, M. T. Oliver, T. Tipton et al., Syncytin-1 modulates placental trophoblast cell proliferation by promoting G1/S transition, Cell Signal, vol.25, pp.1027-1062, 2013.

S. T. Chelbi and D. Vaiman, Genetic and epigenetic factors contribute to the onset of preeclampsia, Mol Cell Endocrinol, vol.282, pp.120-129, 2008.

B. Novakovic and R. Saffery, The ever growing complexity of placental epigenetics-role in adverse pregnancy outcomes and fetal programming, Placenta, vol.33, pp.959-70, 2012.

E. C. Nelissen, A. P. Van-montfoort, J. C. Dumoulin, and J. L. Evers, Epigenetics and the placenta, Hum Reprod Update, vol.17, pp.397-417, 2011.

W. Reik and J. Walter, Genomic imprinting: parental influence on the genome, Nat Rev Genet, vol.2, pp.21-32, 2001.

J. F. Wilkins and D. Haig, What good is genomic imprinting: the function of parent-specific gene expression, Nat Rev Genet, vol.4, pp.359-68, 2003.

M. B. Renfree, T. A. Hore, G. Shaw, J. A. Graves, and A. J. Pask, Evolution of genomic imprinting: insights from marsupials and monotremes, Annu Rev Genomics Hum Genet, vol.10, pp.241-62, 2009.

S. Suzuki, R. Ono, T. Narita, A. J. Pask, G. Shaw et al., Retrotransposon silencing by DNA methylation can drive mammalian genomic imprinting, PLoS Genet, vol.3, p.55, 2007.

S. Barbaux, G. Gascoin-lachambre, C. Buffat, P. Monnier, F. Mondon et al., A genome-wide approach reveals novel imprinted genes expressed in the human placenta, Epigenetics, vol.7, pp.1079-90, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01000763

C. Proudhon, R. Duffie, S. Ajjan, M. Cowley, J. Iranzo et al., Protection against de novo methylation is instrumental in maintaining parent-of-origin methylation inherited from the gametes, Mol Cell, vol.47, pp.909-929, 2012.

F. Court, C. Tayama, V. Romanelli, A. Martin-trujillo, I. Iglesias-platas et al.,

, Genome-wide parent-of-origin DNA methylation analysis reveals the intricacies of human imprinting and suggests a germline methylation-independent mechanism of establishment, Genome Res, vol.24, pp.554-69, 2014.

E. Angiolini, A. Fowden, P. Coan, I. Sandovici, P. Smith et al., Regulation of placental efficiency for nutrient transport by imprinted genes, Placenta, vol.27, pp.98-102, 2006.

A. C. Ferguson-smith, T. Moore, J. Detmar, A. Lewis, M. Hemberger et al., Epigenetics and imprinting of the trophoblast-a workshop report, Placenta, vol.27, pp.122-128, 2006.

D. M. Messerschmidt, Should I, stay or should I go: protection and maintenance of DNA methylation at imprinted genes, Epigenetics, vol.7, pp.969-75, 2012.

A. L. Fowden, P. M. Coan, E. Angiolini, G. J. Burton, and C. M. , Imprinted genes and the epigenetic regulation of placental phenotype, Prog Biophys Mol Biol, vol.106, pp.281-289, 2011.

S. Varmuza and K. Miri, What does genetics tell us about imprinting and the placenta connection?, Cell Mol Life Sci, vol.72, pp.51-72, 2015.

F. Rahnama, F. Shafiei, P. D. Gluckman, M. D. Mitchell, and P. E. Lobie, Epigenetic regulation of human trophoblastic cell migration and invasion, Endocrinology, vol.147, pp.5275-83, 2006.

L. Serman, M. Vlahovic, M. Sijan, F. Bulic-jakus, A. Serman et al., The impact of 5-azacytidine on placental weight, glycoprotein pattern and proliferating cell nuclear antigen expression in rat placenta, Placenta, vol.28, pp.803-814, 2007.

M. Constancia, M. Hemberger, J. Hughes, W. Dean, A. Ferguson-smith et al., Placental-specific Igf-Ii Is a major modulator of placental and fetal growth, Nature, vol.417, pp.945-953, 2002.

L. J. Yin, Y. Zhang, P. P. Lv, W. H. He, Y. T. Wu et al., Insufficient maintenance DNA methylation is associated with abnormal embryonic development, BMC Med, vol.10, p.26, 2012.

C. E. Banister, D. C. Koestler, M. A. Maccani, J. F. Padbury, E. A. Houseman et al., Infant growth restriction is associated with distinct patterns of DNA methylation in human placentas, Epigenetics, vol.6, pp.920-927, 2011.

M. Salas, R. John, A. Saxena, S. Barton, D. Frank et al., Placental growth retardation due to loss of imprinting of Phlda2, Mech Dev, vol.121, pp.1199-210, 2004.

A. I. Diplas, L. Lambertini, M. J. Lee, R. Sperling, Y. L. Lee et al., Differential expression of imprinted genes in normal and Iugr human placentas, Epigenetics, vol.4, pp.235-275, 2009.

N. Kumar, J. Leverence, D. Bick, and V. Sampath, Ontogeny of growth-regulating genes in the placenta, Mol Hum Reprod, vol.33, pp.697-708, 2012.

Z. Khalkhali-ellis, Maspin: the new frontier, Clin Cancer Res, vol.12, pp.7279-83, 2006.

A. Dokras, L. M. Gardner, D. A. Kirschmann, E. A. Seftor, and M. J. Hendrix, The tumour suppressor gene maspin is differentially regulated in cytotrophoblasts during human placental development, Placenta, vol.23, pp.274-80, 2002.

S. T. Chelbi, F. Mondon, H. Jammes, C. Buffat, T. M. Mignot et al., Expressional and epigenetic alterations of placental serine protease inhibitors: serpina3 is a potential marker of preeclampsia, Hypertension, vol.49, pp.76-83, 2007.

S. T. Chelbi, M. L. Wilson, A. C. Veillard, S. A. Ingles, J. Zhang et al., Genetic and epigenetic mechanisms collaborate to control serpina3 expression and its association with placental diseases, Hum Mol Genet, vol.21, pp.1968-78, 2012.

Q. Huang, H. Chen, J. Li, M. Oliver, X. Ma et al., Epigenetic and nonepigenetic regulation of syncytin-1 expression in human placenta and cancer tissues, Cell Signal, vol.26, pp.648-56, 2014.

L. Yu, M. Chen, D. Zhao, P. Yi, L. Lu et al., The H19 gene imprinting in normal pregnancy and pre-eclampsia, Placenta, vol.30, pp.443-450, 2009.

L. Anton, A. G. Brown, M. S. Bartolomei, and M. A. Elovitz, Differential methylation of genes associated with cell adhesion in preeclamptic placentas, PLoS One, vol.9, p.100148, 2014.

G. Lazaraviciute, M. Kauser, S. Bhattacharya, and P. Haggarty, A systematic review and meta-analysis of dna methylation levels and imprinting disorders in children conceived by Ivf/Icsi compared with children conceived spontaneously, Hum Reprod Update, vol.20, issue.6, pp.840-52, 2014.

J. P. Vermeiden and R. E. Bernardus, Are imprinting disorders more prevalent after human in vitro fertilization or intracytoplasmic sperm injection?, Fertil Steril, vol.99, pp.642-51, 2013.

A. P. Van-montfoort, L. L. Hanssen, P. De-sutter, S. Viville, J. P. Geraedts et al., Assisted reproduction treatment and epigenetic inheritance, Hum Reprod Update, vol.18, pp.171-97, 2012.

A. S. Doherty, M. R. Mann, K. D. Tremblay, M. S. Bartolomei, and R. M. Schultz, Differential effects of culture on imprinted H19 expression in the preimplantation mouse embryo, Biol Reprod, vol.62, pp.1526-1561, 2000.

P. Fauque, P. Jouannet, C. Lesaffre, M. A. Ripoche, L. Dandolo et al., Assisted reproductive technology affects developmental kinetics, H19 imprinting control region methylation and H19 gene expression in individual mouse embryos, BMC Dev Biol, vol.7, p.116, 2007.

X. W. Liang, X. S. Cui, S. C. Sun, Y. X. Jin, Y. T. Heo et al., Superovulation induces defective methylation in line-1 retrotransposon elements in blastocyst, Reprod Biol Endocrinol, vol.11, p.69, 2013.

M. R. Mann, S. S. Lee, A. S. Doherty, R. I. Verona, L. D. Nolen et al., Selective loss of imprinting in the placenta following preimplantation development in culture, Development, vol.131, pp.3727-3762, 2004.

B. A. Market-velker, A. D. Fernandes, and M. R. Mann, Side-by-side comparison of five commercial media systems in a mouse model: suboptimal in vitro culture interferes with imprint maintenance, Biol Reprod, vol.83, pp.938-50, 2010.

B. A. Market-velker, L. Zhang, L. S. Magri, A. C. Bonvissuto, and M. R. Mann, Dual effects of superovulation: loss of maternal and paternal imprinted methylation in a dose-dependent manner, Hum Mol Genet, vol.19, pp.36-51, 2010.

R. M. Rivera, P. Stein, J. R. Weaver, J. Mager, R. M. Schultz et al., Manipulations of mouse embryos prior to implantation result in aberrant expression of imprinted genes on day 9.5 of development, Hum Mol Genet, vol.17, pp.1-14, 2008.

S. Khosla, W. Dean, D. Brown, W. Reik, and R. Feil, Culture of preimplantation mouse embryos affects fetal development and the expression of imprinted genes, Biol Reprod, vol.64, pp.918-944, 2001.

E. De-waal, W. Mak, S. Calhoun, P. Stein, T. Ord et al., In vitro culture increases the frequency of stochastic epigenetic errors at imprinted genes in placental tissues from mouse concepti produced through assisted reproductive technologies, Biol Reprod, vol.90, p.22, 2014.

P. Fauque, F. Mondon, F. Letourneur, M. A. Ripoche, L. Journot et al., In vitro fertilization and embryo culture strongly impact the placental transcriptome in the mouse model, PLoS One, vol.5, p.9218, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01788865

M. M. Hossain, D. Tesfaye, D. Salilew-wondim, E. Held, M. J. Proll et al., Massive deregulation of Mirnas from nuclear reprogramming errors during trophoblast differentiation for placentogenesis in cloned pregnancy, BMC Genomics, vol.15, p.43, 2014.

S. Katari, N. Turan, M. Bibikova, O. Erinle, R. Chalian et al., DNA methylation and gene expression differences in children conceived in vitro or in vivo, Hum Mol Genet, vol.18, pp.3769-78, 2009.

S. Yamagishi, T. Matsui, K. Nakamura, T. Yoshida, K. Shimizu et al., Pigment-epithelium-derived factor (Pedf) inhibits angiotensin-ii-induced vascular endothelial growth factor (Vegf) expression in molt-3 T cells through anti-oxidative properties, Microvasc Res, vol.71, pp.222-228, 2006.

A. L. Fortier, F. L. Lopes, N. Darricarrere, J. Martel, and J. M. Trasler, Superovulation alters the expression of imprinted genes in the midgestation mouse placenta, Hum Mol Genet, vol.17, pp.1653-65, 2008.

H. Niwa, Y. Toyooka, D. Shimosato, D. Strumpf, K. Takahashi et al., Interaction between Oct3/4 and Cdx2 determines trophectoderm differentiation, Cell, vol.123, pp.917-946, 2005.

G. Giritharan, L. Delle-piane, A. Donjacour, F. J. Esteban, J. A. Horcajadas et al., In vitro culture of mouse embryos reduces differential gene expression between inner cell mass and trophectoderm, Reprod Sci, vol.19, pp.243-52, 2012.

G. Giritharan, S. Talbi, A. Donjacour, D. Sebastiano, F. Dobson et al., Effect of in vitro fertilization on gene expression and development of mouse preimplantation embryos, Reproduction, vol.134, pp.63-72, 2007.

D. Bourc'his, G. L. Xu, C. S. Lin, B. Bollman, and T. H. Bestor, Dnmt3l and the establishment of maternal genomic imprints, Science, vol.294, pp.2536-2545, 2001.

E. Mercier, G. Lissalde-lavigne, and J. C. Gris, Jak2 V617f mutation in unexplained loss of first pregnancy, N Engl J Med, vol.357, pp.1984-1989, 2007.

M. Vatin, S. Bouvier, L. Bellazi, X. Montagutelli, P. Laissue et al., Polymorphisms of human placental alkaline phosphatase are associated with in vitro fertilization success and recurrent pregnancy loss, Am J Pathol, vol.184, pp.362-370, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01943648

N. Pereza, S. Ostojic, M. Volk, M. Kapovic, and B. Peterlin, Matrix metalloproteinases 1, 2, 3 and 9 functional single-nucleotide polymorphisms in idiopathic recurrent spontaneous abortion, Reprod Biomed Online, vol.24, pp.567-75, 2012.

D. Haouzi, S. Assou, C. Dechanet, T. Anahory, H. Dechaud et al., Controlled ovarian hyperstimulation for in vitro fertilization alters endometrial receptivity in humans: protocol effects, Biol Reprod, vol.82, pp.679-86, 2010.

J. A. Horcajadas, A. Riesewijk, J. Polman, R. Van-os, A. Pellicer et al., Effect of controlled ovarian hyperstimulation in Ivf on endometrial gene expression profiles, Mol Hum Reprod, vol.11, pp.195-205, 2005.

G. Ertzeid and R. Storeng, The impact of ovarian stimulation on implantation and fetal development in mice, Hum Reprod, vol.16, pp.221-226, 2001.

J. Farhi, A. Ben-haroush, N. Andrawus, H. Pinkas, O. Sapir et al., High serum oestradiol concentrations in Ivf cycles increase the risk of pregnancy complications related to abnormal placentation, Reprod Biomed Online, vol.21, pp.331-338, 2010.

A. N. Imudia, A. O. Awonuga, J. O. Doyle, A. J. Kaimal, D. L. Wright et al., Peak serum estradiol level during controlled ovarian hyperstimulation is associated with increased risk of small for gestational age and preeclampsia in singleton pregnancies after in vitro fertilization, Fertil Steril, vol.97, pp.1374-1383, 2012.

M. J. Pelinck, M. H. Keizer, A. Hoek, A. H. Simons, K. Schelling et al., Perinatal outcome in singletons after modified natural cycle Ivf and standard Ivf with ovarian stimulation, Eur J Obstet Gynecol Reprod Biol, vol.148, pp.56-61, 2010.

X. L. Hu, C. Feng, X. H. Lin, Z. X. Zhong, Y. M. Zhu et al., High maternal serum estradiol environment in the first trimester is associated with the increased risk of small-for-gestational-age birth, J Clin Endocrinol Metab, vol.99, pp.2217-2241, 2014.

M. A. Mainigi, D. Olalere, I. Burd, C. Sapienza, M. Bartolomei et al., Peri-implantation hormonal milieu: elucidating mechanisms of abnormal placentation and fetal growth, Biol Reprod, vol.90, p.26, 2014.

A. K. Henningsen, A. Pinborg, O. Lidegaard, C. Vestergaard, J. L. Forman et al., Perinatal outcome of singleton siblings born after assisted reproductive technology and spontaneous conception: Danish national sibling-cohort study, Fertil Steril, vol.95, pp.959-63, 2011.

S. Pelkonen, R. Koivunen, M. Gissler, S. Nuojua-huttunen, A. M. Suikkari et al., Perinatal outcome of children born after frozen and fresh embryo transfer: the finnish cohort study, Hum Reprod, vol.25, pp.914-937, 1995.

L. B. Romundstad, P. R. Romundstad, A. Sunde, V. Von-during, R. Skjaerven et al., Effects of technology or maternal factors on perinatal outcome after assisted fertilisation: a population-based cohort study, Lancet, vol.372, pp.737-780, 2008.

A. Pinborg, A. Loft, A. Henningsen, A. K. Rasmussen, S. Andersen et al., Infant outcome of 957 singletons born after frozen embryo replacement: the Danish National Cohort Study, Fertil Steril, vol.94, pp.1320-1327, 1995.

E. C. Nelissen, J. C. Dumoulin, A. Daunay, J. L. Evers, J. Tost et al., Placentas from pregnancies conceived by Ivf/Icsi have a reduced dna methylation level at the H19 and mest differentially methylated regions, Hum Reprod, vol.28, pp.1117-1143, 2013.

N. Turan, S. Katari, L. F. Gerson, R. Chalian, M. W. Foster et al., Inter-and intra-individual variation in allele-specific DNA methylation and gene expression in children conceived using assisted reproductive technology, PLoS Genet, vol.6, p.1001033, 2010.

U. Zechner, G. Pliushch, E. Schneider, E. Hajj, N. Tresch et al., Quantitative methylation analysis of developmentally important genes in human pregnancy losses after art and spontaneous conception, Mol Hum Reprod, vol.16, pp.704-717, 2010.

R. K. Yuen, L. Avila, M. S. Penaherrera, P. Von-dadelszen, L. Lefebvre et al., Human placental-specific epipolymorphism and its association with adverse pregnancy outcomes, PLoS One, vol.4, p.7389, 2009.

D. J. Barker, The origins of the developmental origins theory, J Intern Med, vol.261, pp.412-419, 2007.

A. R. Isles and A. J. Holland, Imprinted genes and mother-offspring Interactions, Early Hum Dev, vol.81, pp.73-80, 2005.

P. W. Nathanielsz, Animal models that elucidate basic principles of the developmental origins of adult diseases, ILAR J, vol.47, pp.73-82, 2006.

L. P. Reynolds and J. S. Caton, Role of the pre-and post-natal environment in developmental programming of health and productivity, Mol Cell Endocrinol, vol.354, pp.54-63, 2012.

M. W. Gillman, Developmental origins of health and disease, N Engl J Med, vol.353, pp.1848-50, 2005.

P. D. Gluckman, M. A. Hanson, C. Cooper, and K. L. Thornburg, Effect of in utero and early-life conditions on adult health and disease, N Engl J Med, vol.359, pp.61-73, 2008.

R. A. Simmons, L. J. Templeton, and S. J. Gertz, Intrauterine growth retardation leads to the development of type 2 diabetes in the rat, Diabetes, vol.50, pp.2279-86, 2001.

R. A. Simmons, Developmental origins of adult disease, Pediatr Clin North Am, vol.56, pp.449-66, 2009.

D. J. Barker, C. Osmond, K. L. Thornburg, E. Kajantie, and J. G. Eriksson, The lifespan of men and the shape of their placental surface at birth, Placenta, vol.32, pp.783-790, 2011.

J. G. Eriksson, E. Kajantie, K. L. Thornburg, C. Osmond, and D. J. Barker, Mother's body size and placental size predict coronary heart disease in men, Eur Heart J, vol.32, pp.2297-303, 2011.

D. J. Barker, A. R. Bull, C. Osmond, and S. J. Simmonds, Fetal and placental size and risk of hypertension in adult life, BMJ, vol.301, pp.259-62, 1990.

R. Hart and R. J. Norman, The longer-term health outcomes for children born as a result of Ivf treatment: part I-general health outcomes, Hum Reprod Update, vol.19, pp.232-275, 2013.

M. Ceelen, M. M. Van-weissenbruch, J. Prein, J. J. Smit, J. P. Vermeiden et al., Growth during infancy and early childhood in relation to blood pressure and body fat measures at age 8-18 years of Ivf children and spontaneously conceived controls born to subfertile parents, Hum Reprod, vol.24, pp.2788-95, 2009.

M. Ceelen, M. M. Van-weissenbruch, J. C. Roos, J. P. Vermeiden, F. E. Van-leeuwen et al., Body composition in children and adolescents born after in vitro fertilization or spontaneous conception, J Clin Endocrinol Metab, vol.92, pp.3417-3440, 2007.

M. Ceelen, M. M. Van-weissenbruch, J. P. Vermeiden, F. E. Van-leeuwen, and H. A. Delemarre-van-de-waal, Growth and development of children born after in vitro fertilization, Fertil Steril, vol.90, pp.1662-73, 2008.

H. L. Miles, P. L. Hofman, J. Peek, M. Harris, D. Wilson et al., In vitro fertilization improves childhood growth and metabolism, J Clin Endocrinol Metab, vol.92, pp.3441-3446, 2007.

S. D. Sakka, D. Loutradis, C. Kanaka-gantenbein, A. Margeli, M. Papastamataki et al., Absence of insulin resistance and low-grade inflammation despite early metabolic syndrome manifestations in children born after in vitro fertilization, Fertil Steril, vol.94, pp.1693-1702, 2010.

B. Portha, A. Fournier, M. D. Kioon, V. Mezger, and J. Movassat, Early environmental factors, alteration of epigenetic marks and metabolic disease susceptibility, Biochimie, vol.97, pp.1-15, 2014.

V. F. Oliver, H. L. Miles, W. S. Cutfield, P. L. Hofman, J. L. Ludgate et al., Defects in imprinting and genome-wide DNA methylation are not common in the in vitro fertilization population, Fertil Steril, vol.97, pp.147-53, 2012.

S. E. Puumala, H. H. Nelson, J. A. Ross, R. H. Nguyen, M. A. Damario et al., Similar DNA methylation levels in specific imprinting control regions in children conceived with and without assisted reproductive technology: a cross-sectional study, BMC Pediatr, vol.12, p.33, 2012.

N. Whitelaw, S. Bhattacharya, G. Hoad, G. W. Horgan, M. Hamilton et al., Epigenetic status in the offspring of spontaneous and assisted conception, Hum Reprod, vol.29, pp.1452-1460, 2014.

E. Rexhaj, A. Paoloni-giacobino, S. F. Rimoldi, D. G. Fuster, M. Anderegg et al., Mice generated by in vitro fertilization exhibit vascular dysfunction and shortened life span, J Clin Invest, vol.123, pp.5052-60, 2013.

E. De-waal, Y. Yamazaki, P. Ingale, M. Bartolomei, R. Yanagimachi et al., Primary epimutations introduced during intracytoplasmic sperm injection (Icsi) are corrected by germline-specific epigenetic reprogramming, Proc Natl Acad Sci U S A, vol.109, pp.4163-4171, 2012.

M. Ruiz-alonso, D. Blesa, P. Diaz-gimeno, E. Gomez, M. Fernandez-sanchez et al., The endometrial receptivity array for diagnosis and personalized embryo transfer as a treatment for patients with repeated implantation failure, Fertil steril, vol.100, pp.818-842, 2013.

A. Maheshwari and S. Bhattacharya, Elective frozen replacement cycles for all: ready for prime time?, Hum Reprod, vol.28, pp.6-9, 2013.

L. Petrussa, H. Van-de-velde, D. Rycke, and M. , Dynamic regulation of DNA methyltransferases in human oocytes and preimplantation embryos after assisted reproductive technologies, Mol Hum Reprod, vol.20, pp.861-74, 2014.

P. D. Gluckman, K. A. Lillycrop, M. H. Vickers, A. B. Pleasants, E. S. Phillips et al., Metabolic plasticity during mammalian development is directionally dependent on early nutritional status, Proc Natl Acad Sci, vol.104, pp.12796-800, 2007.

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, pp.531-540, 2003.

H. Wang and S. K. Dey, Roadmap to embryo implantation: clues from mouse models, Nat Rev Genet, vol.7, pp.185-99, 2006.

J. Rossant and J. C. Cross, Placental development: lessons from mouse mutants, Nat Rev Genet, vol.2, pp.538-586, 2001.

O. Basso, C. R. Weinberg, D. D. Baird, A. J. Wilcox, and J. Olsen, Subfecundity as a correlate of preeclampsia: a study within the Danish National Birth Cohort, Am J Epidemiol, vol.157, pp.195-202, 2003.

Z. Pandian, S. Bhattacharya, and A. Templeton, Review of unexplained infertility and obstetric outcome: a 10 year review, Hum Reprod, vol.16, pp.2593-2600, 2001.

B. Horsthemke and M. Ludwig, Assisted reproduction: the epigenetic perspective, Hum Reprod Update, vol.11, pp.473-82, 2005.

B. C. Christensen, E. A. Houseman, C. J. Marsit, S. Zheng, M. R. Wrensch et al., Aging and environmental exposures alter tissue-specific DNA methylation dependent upon Cpg island context, PLoS Genet, vol.5, p.1000602, 2009.

D. I. Schroeder, J. D. Blair, P. Lott, Y. Ho, D. Hong et al., The human placenta methylome, Proc Natl Acad Sci U S A, vol.110, pp.6037-6079, 2013.

F. R. Wu, Y. Liu, M. B. Shang, X. X. Yang, B. Ding et al., Differences in H3k4 trimethylation in in vivo and in vitro fertilization mouse preimplantation embryos, Genet Mol Res, vol.11, pp.1099-108, 2012.

E. M. Rosenbluth, D. N. Shelton, A. E. Sparks, E. Devor, L. Christenson et al., Microrna expression in the human blastocyst, Fertil Steril, vol.99, pp.855-61, 2013.

V. L. Clifton, Review: sex and the human placenta: mediating differential strategies of fetal growth and survival, Placenta, vol.31, pp.33-42, 2010.

D. Feil, M. Lane, C. T. Roberts, R. L. Kelley, L. J. Edwards et al., Effect of culturing mouse embryos under different oxygen concentrations on subsequent fetal and placental development, J Physiol, vol.572, pp.87-96, 2006.

Z. Wang, L. Xu, and F. He, Embryo vitrification affects the methylation of the H19/Igf2 differentially methylated domain and the expression of H19 and Igf2, Fertil Steril, vol.93, pp.2729-2762, 2010.

M. V. Gomes, J. Huber, R. A. Ferriani, A. Neto, A. M. Ramos et al., Abnormal methylation at the Kvdmr1 imprinting control region in clinically normal children conceived by assisted reproductive technologies, Mol Hum Reprod, vol.15, pp.471-478, 2009.

. Choux, Clinical Epigenetics, vol.7, p.87, 2015.

R. Barker and D. J. , The origins of the developmental origins theory, J Intern Med, vol.261, pp.412-417, 2007.

J. Boivin, L. Bunting, J. A. Collins, and K. G. Nygren, International estimates of infertility prevalence and treatment-seeking: potential need and demand for infertility medical care, Hum Reprod, vol.22, pp.1506-1512, 2007.

P. A. Bolze, M. Mommert, and F. Mallet, Contribution of syncytins and other endogenous retroviral envelopes to human placenta pathologies, Prog Mol Biol Transl Sci, vol.145, pp.111-162, 2017.

C. Bruno, V. Carmignac, I. Netchine, C. Choux, Y. Duffourd et al., Germline correction of an epimutation related to Silver-Russell syndrome, Hum Mol Genet, vol.24, pp.3314-3321, 2015.

C. Camprubi, I. Iglesias-platas, A. Martin-trujillo, C. Salvador-alarcon, M. A. Rodriguez et al., Stability of genomic imprinting and gestational-age dynamic methylation in complicated pregnancies conceived following assisted reproductive technologies, Biol Reprod, vol.89, p.50, 2013.

C. Choux, V. Carmignac, C. Bruno, P. Sagot, D. Vaiman et al., The placenta: phenotypic and epigenetic modifications induced by Assisted Reproductive Technologies throughout pregnancy, Clin Epigenetics, vol.7, p.87, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01188079

M. S. Comeaux, A. M. Roy-engel, D. J. Hedges, and P. L. Deininger, Diverse cis factors controlling Alu retrotransposition: what causes Alu elements to die?, Genome Res, vol.19, pp.545-555, 2009.

E. De-waal, W. Mak, S. Calhoun, P. Stein, T. Ord et al., In vitro culture increases the frequency of stochastic epigenetic errors at imprinted genes

, Biol Reprod, vol.90, p.22, 2014.

E. De-waal, L. A. Vrooman, E. Fischer, T. Ord, M. A. Mainigi et al., The cumulative effect of assisted reproduction procedures on placental development and epigenetic perturbations in a mouse model, Hum Mol Genet, vol.24, pp.6975-6985, 2015.

L. Desch, C. Bruno, M. Luu, J. Barberet, C. Choux et al., Embryo multinucleation at the two-cell stage is an independent predictor of intracytoplasmic sperm injection outcomes, Fertil Steril, vol.107, pp.97-103, 2017.

P. Fauque and D. Bourc'his, Genes are not the whole story: retrotransposons as new determinants of male fertility, Textbook of Human Reproductive Genetics, pp.83-96, 2014.

P. Fauque, M. A. Ripoche, J. Tost, L. Journot, A. Gabory et al., Modulation of imprinted gene network in placenta results in normal development of in vitro manipulated mouse embryos, Hum Mol Genet, vol.19, pp.1779-1790, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01788884

A. L. Fortier, F. L. Lopes, N. Darricarrere, J. Martel, and J. M. Trasler, Superovulation alters the expression of imprinted genes in the midgestation mouse placenta, Hum Mol Genet, vol.17, pp.1653-1665, 2008.

A. L. Fortier, S. Mcgraw, F. L. Lopes, K. M. Niles, M. Landry et al., Modulation of imprinted gene expression following superovulation, Mol Cell Endocrinol, vol.388, pp.51-57, 2014.

M. Hansen, C. Bower, E. Milne, D. Klerk, N. Kurinczuk et al., Assisted reproductive technologies and the risk of birth defects-a systematic review, Hum Reprod, vol.20, pp.328-338, 2005.

Z. M. He, J. Li, Y. L. Hwa, B. Brost, Q. Fang et al., Transition of LINE-1 DNA methylation status and altered expression in first and third trimester placentas, PLoS One, vol.9, p.96994, 2014.

K. Hogg, E. M. Price, and W. P. Robinson, Improved reporting of DNA methylation data derived from studies of the human placenta, Epigenetics, vol.9, pp.333-337, 2014.

T. Hu, X. Zhu, W. Pi, M. Yu, H. Shi et al., Hypermethylated LTR retrotransposon exhibits enhancer activity, Epigenetics, vol.12, pp.226-237, 2017.

M. Hyrapetian, E. M. Loucaides, and A. G. Sutcliffe, Health and disease in children born after assistive reproductive therapies (ART), J Reprod Immunol, vol.106, pp.21-26, 2014.

I. Iglesias-platas, A. Martin-trujillo, P. Petazzi, A. Guillaumet-adkins, M. Esteller et al., Altered expression of the imprinted transcription factor PLAGL1 deregulates a network of genes in the human IUGR placenta, Hum Mol Genet, vol.23, pp.6275-6285, 2014.

H. Kano, I. Godoy, C. Courtney, M. R. Vetter, G. L. Gerton et al., L1 retrotransposition occurs mainly in embryogenesis and creates somatic mosaicism, Genes Dev, vol.23, pp.1303-1312, 2009.

H. R. Kohan-ghadr, L. Kadam, C. Jain, D. R. Armant, and S. Drewlo, Potential role of epigenetic mechanisms in regulation of trophoblast differentiation, migration, and invasion in the human placenta, Cell Adh Migr, vol.10, pp.126-135, 2016.

G. Lazaraviciute, M. Kauser, S. Bhattacharya, and P. Haggarty, A systematic review and meta-analysis of DNA methylation levels and imprinting disorders in children conceived by IVF/ICSI compared with children conceived spontaneously, Hum Reprod Update, vol.20, pp.840-852, 2014.

J. F. Litzky, M. A. Deyssenroth, T. M. Everson, D. A. Armstrong, L. Lambertini et al., Placental imprinting variation associated with assisted reproductive technologies and subfertility, Epigenetics, vol.12, pp.653-661, 2017.

A. G. Lokossou, C. Toudic, and B. Barbeau, Implication of human endogenous retrovirus envelope proteins in placental functions, Viruses, vol.6, pp.4609-4627, 2014.

N. Melamed, S. Choufani, L. E. Wilkins-haug, G. Koren, and R. Weksberg, Comparison of genome-wide and gene-specific DNA methylation between ART and naturally conceived pregnancies, Epigenetics, vol.10, pp.474-483, 2015.

D. Monk, J. Morales, J. T. Den-dunnen, S. Russo, F. Court et al., Recommendations for a nomenclature system for reporting methylation aberrations in imprinted domains, Epigenetics, p.0, 2016.

G. E. Moore, M. Ishida, C. Demetriou, L. Al-olabi, L. J. Leon et al., The role and interaction of imprinted genes in human fetal growth, Philos Trans R Soc Lond B Biol Sci, vol.370, 2015.

E. C. Nelissen, J. C. Dumoulin, A. Daunay, J. L. Evers, J. Tost et al., Placentas from pregnancies conceived by IVF/ICSI have a reduced DNA methylation level at the H19 and MEST differentially methylated regions, Hum Reprod, vol.28, pp.1117-1126, 2013.

S. Noorali, I. C. Rotar, C. Lewis, J. P. Pestaner, D. G. Pace et al., Role of HERV-W syncytin-1 in placentation and maintenance of human pregnancy, Appl Immunohistochem Mol Morphol, vol.17, pp.319-328, 2009.

A. Pinborg, U. B. Wennerholm, L. B. Romundstad, A. Loft, K. Aittomaki et al., Why do singletons conceived after assisted reproduction technology have adverse perinatal outcome? Systematic review and meta-analysis, Hum Reprod Update, vol.19, pp.87-104, 2013.

J. Qin, X. Liu, X. Sheng, H. Wang, and S. Gao, Assisted reproductive technology and the risk of pregnancy-related complications and adverse pregnancy outcomes in singleton pregnancies: a meta-analysis of cohort studies, Fertil Steril, vol.105, pp.73-85, 2016.

R. C. Rancourt, H. R. Harris, and K. B. Michels, Methylation levels at imprinting control regions are not altered with ovulation induction or in vitro fertilization in a birth cohort, Hum Reprod, vol.27, pp.2208-2216, 2012.

T. Rousseau, C. Ferdynus, C. Quantin, J. B. Gouyon, and P. Sagot, Liveborn birthweight of single and uncomplicated pregnancies between 28 and 42 weeks of gestation from Burgundy perinatal network

, J Gynecol Obstet Biol Reprod (Paris), vol.37, pp.589-596, 2008.

S. Sakian, K. Louie, E. C. Wong, J. Havelock, S. Kashyap et al., Altered gene expression of H19 and IGF2 in placentas from ART pregnancies, Placenta, vol.36, pp.1100-1105, 2015.

X. Shi, Y. Ni, H. Zheng, S. Chen, M. Zhong et al., Abnormal methylation patterns at the IGF2/H19 imprinting control region in phenotypically normal babies conceived by assisted reproductive technologies, Eur J Obstet Gynecol Reprod Biol, vol.158, pp.52-55, 2011.

J. M. Thompson, L. M. Irgens, R. Skjaerven, and S. Rasmussen, Placenta weight percentile curves for singleton deliveries, BJOG, vol.114, pp.715-720, 2007.

S. Tierling, N. Y. Souren, J. Gries, C. Loporto, M. Groth et al., Assisted reproductive technologies do not enhance the variability of DNA methylation imprints in human, J Med Genet, vol.47, pp.371-376, 2010.

J. M. Tolosa, J. E. Schjenken, V. L. Clifton, A. Vargas, B. Barbeau et al., The endogenous retroviral envelope protein syncytin-1 inhibits LPS/PHA-stimulated cytokine responses in human blood and is sorted into placental exosomes, Placenta, vol.33, pp.933-941, 2012.

P. , C. C. Sapienza, and C. , Inter-and intra-individual variation in allele-specific DNA methylation and gene expression in children conceived using assisted reproductive technology, PLoS Genet, vol.6, p.1001033, 2010.

J. A. Van-den-hurk, I. C. Meij, M. C. Seleme, H. Kano, K. Nikopoulos et al., L 1 retrotransposition can occur early in human embryonic development, Hum Mol Genet, vol.16, pp.1587-1592, 2007.

S. J. Van-dijk, J. Zhou, T. J. Peters, M. Buckley, B. Sutcliffe et al., Effect of prenatal DHA supplementation on the infant epigenome: results from a randomized controlled trial, Clin Epigenetics, vol.8, p.114, 2016.

A. Vargas, S. Zhou, M. Ethier-chiasson, D. Flipo, J. Lafond et al., Syncytin proteins incorporated in placenta exosomes are important for cell uptake and show variation in abundance in serum exosomes from patients with preeclampsia, FASEB J, vol.28, pp.3703-3719, 2014.

A. Varrault, C. Gueydan, A. Delalbre, A. Bellmann, S. Houssami et al., Zac1 regulates an imprinted gene network critically involved in the control of embryonic growth, Dev Cell, vol.11, pp.711-722, 2006.
URL : https://hal.archives-ouvertes.fr/inserm-00158399

R. N. Vincent, L. D. Gooding, K. Louie, C. Wong, E. Ma et al., Altered DNA methylation and expression of PLAGL1 in cord blood from assisted reproductive technology pregnancies compared with natural conceptions, Fertil Steril, vol.106, pp.739-748, 2016.

Z. Wang, L. Xu, and F. He, Embryo vitrification affects the methylation of the H19/Igf2 differentially methylated domain and the expression of H19 and Igf2, Fertil Steril, vol.93, pp.2729-2733, 2010.

E. C. Wong, C. Hatakeyama, W. P. Robinson, and S. Ma, DNA methylation at H19/IGF2 ICR1 in the placenta of pregnancies conceived by in vitro fertilization and intracytoplasmic sperm injection, Fertil Steril, vol.95, pp.2524-2526, 2011.

R. K. Yuen and W. P. Robinson, Review: a high capacity of the human placenta for genetic and epigenetic variation: implications for assessing pregnancy outcome, Placenta, vol.32, pp.136-141, 2011.

U. Zechner, G. Pliushch, E. Schneider, E. Hajj, N. Tresch et al., Quantitative methylation analysis of developmentally important genes in human pregnancy losses after ART and spontaneous conception, Mol Hum Reprod, vol.16, pp.704-713, 2010.

L. Zhu, Y. Zhang, Y. Liu, R. Zhang, Y. Wu et al., Maternal and live-birth outcomes of pregnancies following assisted reproductive technology: a retrospective cohort study, Sci Rep, vol.6, p.35141, 2016.

C. Messerlian and A. J. Gaskins, Epidemiologic Approaches for Studying Assisted Reproductive Technologies: Design, Methods, Analysis and Interpretation, Curr Epidemiol Rep, issue.4, pp.124-132, 2017.

G. Lazaraviciute, M. Kauser, S. Bhattacharya, and P. Haggarty, A systematic review and metaanalysis of DNA methylation levels and imprinting disorders in children conceived by IVF/ICSI compared with children conceived spontaneously, Hum Reprod Update, issue.20, pp.840-52, 2014.

A. Pinborg, U. B. Wennerholm, L. B. Romundstad, A. Loft, K. Aittomaki et al., Why do singletons conceived after assisted reproduction technology have adverse perinatal outcome? Systematic review and meta-analysis, Hum Reprod Update, issue.19, pp.87-104, 2013.

J. Qin, X. Liu, X. Sheng, H. Wang, and S. Gao, Assisted reproductive technology and the risk of pregnancy-related complications and adverse pregnancy outcomes in singleton pregnancies: a meta-analysis of cohort studies, Fertil Steril, issue.105, pp.73-85, 2016.

L. Zhu, Y. Zhang, Y. Liu, R. Zhang, Y. Wu et al., Maternal and Live-birth Outcomes of Pregnancies following Assisted Reproductive Technology: A Retrospective Cohort Study, Sci Rep, issue.6, p.35141, 2016.

C. Choux, V. Carmignac, C. Bruno, P. Sagot, D. Vaiman et al., The placenta: phenotypic and epigenetic modifications induced by Assisted Reproductive Technologies throughout pregnancy, Clin Epigenetics, issue.7, p.87, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01188079

E. Angiolini, A. Fowden, P. Coan, I. Sandovici, P. Smith et al., Regulation of placental efficiency for nutrient transport by imprinted genes, Placenta, issue.27, pp.98-102, 2006.

A. C. Ferguson-smith, T. Moore, J. Detmar, A. Lewis, M. Hemberger et al., Epigenetics and imprinting of the trophoblast --a workshop report, Placenta, issue.27, pp.122-128, 2006.

D. Monk, Genomic imprinting in the human placenta, Am J Obstet Gynecol, issue.213, pp.152-62, 2015.

B. Portha, A. Fournier, M. D. Kioon, V. Mezger, and M. J. , Early environmental factors, alteration of epigenetic marks and metabolic disease susceptibility, Biochimie, issue.97, pp.1-15, 2014.

P. J. Rugg-gunn, Epigenetic features of the mouse trophoblast, Reprod Biomed Online, issue.25, pp.21-30, 2012.

H. R. Kohan-ghadr, L. Kadam, C. Jain, D. R. Armant, and S. Drewlo, Potential role of epigenetic mechanisms in regulation of trophoblast differentiation, migration, and invasion in the human placenta, Cell Adh Migr, issue.10, pp.126-161, 2016.

A. C. Eddy, H. Chapman, and G. E. , Acute Hypoxia and Chronic Ischemia Induce Differential Total Changes in Placental Epigenetic Modifications, Reprod Sci, 2018.

B. Rahat, A. Mahajan, R. Bagga, A. Hamid, and J. Kaur, Epigenetic modifications at DMRs of placental genes are subjected to variations in normal gestation, pathological conditions and folate supplementation, Sci Rep, issue.7, p.40774, 2017.

R. Barouki, E. Melen, Z. Herceg, J. Beckers, J. Chen et al., Epigenetics as a mechanism linking developmental exposures to long-term toxicity, Environ Int, issue.114, pp.77-86, 2018.

C. Choux, C. Binquet, V. Carmignac, C. Bruno, C. Chapusot et al., The epigenetic control of transposable elements and imprinted genes in newborns is affected by the mode of conception: ART versus spontaneous conception without underlying infertility, Hum Reprod, issue.33, pp.331-340, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01689456

D. Monk, J. Morales, J. T. Den-dunnen, S. Russo, F. Court et al., Recommendations for a nomenclature system for reporting methylation aberrations in imprinted domains, Epigenetics, issue.13, pp.117-121, 2018.

D. Umlauf, Y. Goto, R. Cao, F. Cerqueira, A. Wagschal et al., Imprinting along the Kcnq1 domain on mouse chromosome 7 involves repressive histone methylation and recruitment of Polycomb group complexes, Nat Genet, issue.36, pp.1296-300, 2004.
URL : https://hal.archives-ouvertes.fr/hal-02262427

M. Jahangiri, M. Shahhoseini, and B. Movaghar, H19 and MEST gene expression and histone modification in blastocysts cultured from vitrified and fresh two-cell mouse embryos, Reprod Biomed Online, issue.29, pp.559-66, 2014.

D. Monk, P. Arnaud, S. Apostolidou, F. A. Hills, G. Kelsey et al., Limited evolutionary conservation of imprinting in the human placenta, Proc Natl Acad Sci, issue.103, pp.6623-6631, 2006.
URL : https://hal.archives-ouvertes.fr/hal-01934548

T. Rousseau, C. Ferdynus, C. Quantin, J. B. Gouyon, and P. Sagot, Liveborn birth-weight of single and uncomplicated pregnancies between 28 and 42 weeks of gestation from Burgundy perinatal network

, J Gynecol Obstet Biol Reprod (Paris), issue.37, pp.589-96, 2008.

J. M. Thompson, L. M. Irgens, R. Skjaerven, and S. Rasmussen, Placenta weight percentile curves for singleton deliveries, BJOG, issue.114, pp.715-735, 2007.

S. Katari, N. Turan, M. Bibikova, O. Erinle, R. Chalian et al., DNA methylation and gene expression differences in children conceived in vitro or in vivo, Hum Mol Genet, issue.18, pp.3769-78, 2009.

E. C. Nelissen, J. C. Dumoulin, A. Daunay, J. L. Evers, J. Tost et al., Placentas from pregnancies conceived by IVF/ICSI have a reduced DNA methylation level at the H19 and MEST differentially methylated regions, Hum Reprod, issue.28, pp.1117-1143, 2013.

R. C. Rancourt, H. R. Harris, and K. B. Michels, Methylation levels at imprinting control regions are not altered with ovulation induction or in vitro fertilization in a birth cohort, Hum Reprod, issue.27, pp.2208-2224, 2012.

C. Camprubi, I. Iglesias-platas, A. Martin-trujillo, C. Salvador-alarcon, M. A. Rodriguez et al., Stability of genomic imprinting and gestational-age dynamic methylation in complicated pregnancies conceived following assisted reproductive technologies, Biol Reprod, issue.89, p.50, 2013.

N. Turan, S. Katari, L. F. Gerson, R. Chalian, M. W. Foster et al., Inter-and intraindividual variation in allele-specific DNA methylation and gene expression in children conceived using assisted reproductive technology, PLoS Genet, issue.6, p.1001033, 2010.

M. Jahangiri, M. Shahhoseini, and B. Movaghar, The Effect of Vitrification on Expression and Histone Marks of Igf2 and Oct4 in Blastocysts Cultured from Two-Cell Mouse Embryos, Cell J, issue.19, pp.607-613, 2018.

T. Li, T. H. Vu, G. A. Ulaner, E. Littman, J. Q. Ling et al., IVF results in de novo DNA methylation and histone methylation at an Igf2-H19 imprinting epigenetic switch, Mol Hum Reprod, issue.11, pp.631-671, 2005.

A. L. Fortier, F. L. Lopes, N. Darricarrere, J. Martel, and J. M. Trasler, Superovulation alters the expression of imprinted genes in the midgestation mouse placenta, Hum Mol Genet, issue.17, pp.1653-65, 2008.

A. L. Fortier, S. Mcgraw, F. L. Lopes, K. M. Niles, M. Landry et al., Modulation of imprinted gene expression following superovulation, Mol Cell Endocrinol, issue.388, pp.51-58, 2014.

G. E. Moore, M. Ishida, C. Demetriou, L. Al-olabi, L. J. Leon et al., The role and interaction of imprinted genes in human fetal growth, Philos Trans R Soc Lond B Biol Sci, issue.370, p.20140074, 2015.

J. F. Litzky, S. L. Boulet, N. Esfandiari, Y. Zhang, D. M. Kissin et al., Effect of frozen/thawed embryo transfer on birthweight, macrosomia, and low birthweight rates in US singleton infants, Am J Obstet Gynecol, vol.433, issue.218, p.10, 2018.

A. Sazonova, K. Kallen, A. Thurin-kjellberg, U. B. Wennerholm, and C. Bergh, Obstetric outcome in singletons after in vitro fertilization with cryopreserved/thawed embryos, Hum Reprod, issue.27, pp.1343-50, 2012.

C. Allen, S. Bowdin, R. F. Harrison, A. G. Sutcliffe, L. Brueton et al., Pregnancy and perinatal outcomes after assisted reproduction: a comparative study, Ir J Med Sci, vol.177, issue.3, pp.233-241, 2008.

D. J. Amor, J. X. Xu, J. L. Halliday, I. Francis, D. L. Healy et al., Pregnancies conceived using assisted reproductive technologies (ART) have low levels of pregnancy-associated plasma protein-A (PAPP-A) leading to a high rate of false-positive results in first trimester screening for Down syndrome, Hum Reprod, vol.24, issue.6, pp.1330-1338, 2009.

M. Ashrafi, R. Gosili, R. Hosseini, A. Arabipoor, J. Ahmadi et al., Risk of gestational diabetes mellitus in patients undergoing assisted reproductive techniques, Eur J Obstet Gynecol Reprod Biol, vol.176, pp.149-152, 2014.

B. O. Asvold, L. J. Vatten, T. G. Tanbo, and A. Eskild, Concentrations of human chorionic gonadotrophin in very early pregnancy and subsequent pre-eclampsia: a cohort study, Hum Reprod, vol.29, issue.6, pp.1153-1160, 2014.

S. Azzi, T. C. Sas, Y. Koudou, L. Bouc, Y. Souberbielle et al., Degree of methylation of ZAC1 (PLAGL1) is associated with prenatal and post-natal growth in healthy infants of the EDEN mother child cohort, Epigenetics, vol.9, issue.3, pp.338-345, 2014.

D. J. Barker, The origins of the developmental origins theory, J Intern Med, vol.261, issue.5, pp.412-417, 2007.

D. J. Barker, C. Osmond, K. L. Thornburg, K. E. Eriksson, and J. G. , The lifespan of men and the shape of their placental surface at birth, Placenta, vol.32, issue.10, pp.783-787, 2011.

R. Barouki, E. Melen, Z. Herceg, J. Beckers, J. Chen et al., Epigenetics as a mechanism linking developmental exposures to long-term toxicity, Environ Int, vol.114, pp.77-86, 2018.

E. Bloise, W. Lin, X. Liu, R. Simbulan, K. S. Kolahi et al., Impaired placental nutrient transport in mice generated by in vitro fertilization, Endocrinology, vol.153, issue.7, pp.3457-3467, 2012.

P. A. Bolze, M. Mommert, and F. Mallet, Contribution of Syncytins and Other Endogenous Retroviral Envelopes to Human Placenta Pathologies, Prog Mol Biol Transl Sci, vol.145, pp.111-162, 2017.

&. Bourc and T. H. Bestor, Meiotic catastrophe and retrotransposon reactivation in male germ cells lacking Dnmt3L, Nature, vol.431, issue.7004, pp.96-99, 2004.

D. K. Bourque, L. Avila, M. Penaherrera, V. Dadelszen, P. et al., Decreased placental methylation at the H19/IGF2 imprinting control region is associated with normotensive intrauterine growth restriction but not preeclampsia, Placenta, vol.31, issue.3, pp.197-202, 2010.

M. Brandes, J. C. Verzijden, C. J. Hamilton, D. Weys, N. P. et al., Is the fertility treatment itself a risk factor for early pregnancy loss?, Reprod Biomed Online, vol.22, issue.2, pp.192-199, 2011.

C. Camprubi, I. Iglesias-platas, A. Martin-trujillo, C. Salvador-alarcon, M. A. Rodriguez et al., Stability of genomic imprinting and gestational-age dynamic methylation in complicated pregnancies conceived following assisted reproductive technologies, Biol Reprod, vol.89, issue.3, p.50, 2013.

D. Caserta, R. Marci, C. Tatone, M. Schimberni, E. Vaquero et al., IVF pregnancies: neonatal outcomes after the new Italian law on assisted reproduction technology (law 40/2004), Acta Obstet Gynecol Scand, vol.87, issue.9, pp.935-939, 2008.

P. Cavoretto, M. Candiani, V. Giorgione, A. Inversetti, M. M. Abu-saba et al., Risk of spontaneous preterm birth in singleton pregnancies conceived after IVF/ICSI treatment: meta-analysis of cohort studies, Ultrasound Obstet Gynecol, vol.51, issue.1, pp.43-53, 2018.

H. Cedar and Y. Bergman, Programming of DNA methylation patterns, Annu Rev Biochem, vol.81, pp.97-117, 2012.

M. Ceelen, M. M. Van-weissenbruch, J. Prein, J. J. Smit, J. P. Vermeiden et al., Growth during infancy and early childhood in relation to blood pressure and body fat measures at age 8-18 years of IVF children and spontaneously conceived controls born to subfertile parents, Hum Reprod, vol.24, issue.11, pp.2788-2795, 2009.

M. Ceelen, M. M. Van-weissenbruch, J. C. Roos, J. P. Vermeiden, F. E. Van-leeuwen et al., Body composition in children and adolescents born after in vitro fertilization or spontaneous conception, J Clin Endocrinol Metab, vol.92, issue.9, pp.3417-3423, 2007.

M. Ceelen, M. M. Van-weissenbruch, J. P. Vermeiden, F. E. Van-leeuwen, D. De-waal et al., Growth and development of children born after in vitro fertilization, Fertil Steril, vol.90, issue.5, pp.1662-1673, 2008.

P. Chaveeva, I. F. Carbone, A. Syngelaki, R. Akolekar, and K. H. Nicolaides, Contribution of method of conception on pregnancy outcome after the 11-13 weeks scan, Fetal Diagn Ther, vol.30, issue.1, pp.9-22, 2011.

C. St and D. Vaiman, Genetic and epigenetic factors contribute to the onset of preeclampsia, Mol Cell Endocrinol, vol.282, issue.1-2, pp.120-129, 2008.

S. L. Chen, X. Y. Shi, H. Y. Zheng, F. R. Wu, and C. Luo, Aberrant DNA methylation of imprinted H19 gene in human preimplantation embryos, Fertil Steril, vol.94, issue.6, pp.2356-2358, 2010.

X. Chen, Y. Huang, H. Huang, Y. Guan, M. Li et al., Effects of superovulation, in vitro fertilization, and oocyte in vitro maturation on imprinted gene Grb10 in mouse blastocysts, Arch Gynecol Obstet, 2018.

Z. Chen, K. M. Robbins, K. D. Wells, and R. M. Rivera, Large offspring syndrome: a bovine model for the human loss-of-imprinting overgrowth syndrome Beckwith-Wiedemann, Epigenetics, vol.8, issue.6, pp.591-601, 2013.

C. Choux, C. Binquet, V. Carmignac, C. Bruno, C. Chapusot et al., The epigenetic control of transposable elements and imprinted genes in newborns is affected by the mode of conception: ART versus spontaneous conception without underlying infertility, Hum Reprod, vol.33, issue.2, pp.331-340, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01689456

C. Choux, V. Carmignac, C. Bruno, P. Sagot, D. Vaiman et al., The placenta: phenotypic and epigenetic modifications induced by Assisted Reproductive Technologies throughout pregnancy, Clin Epigenetics, vol.7, issue.1, p.87, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01188079

G. A. Clarke and . History, Hum Reprod, vol.21, issue.7, pp.1645-1650, 2006.

C. Kp and V. L. Baker, Corpus luteal contribution to maternal pregnancy physiology and outcomes in assisted reproductive technologies, Am J Physiol Regul Integr Comp Physiol, vol.304, issue.2, pp.69-72, 2013.

D. Kretzer, D. Dennis, P. Hudson, B. Leeton, J. Lopata et al., Transfer of a human zygote, Lancet, vol.2, issue.7831, pp.728-729, 1973.

E. De-waal, W. Mak, S. Calhoun, P. Stein, T. Ord et al., In vitro culture increases the frequency of stochastic epigenetic errors at imprinted genes in placental tissues from mouse concepti produced through assisted reproductive technologies, Biol Reprod, vol.90, issue.2, p.22, 2014.

E. De-waal, L. A. Vrooman, E. Fischer, T. Ord, M. A. Mainigi et al., The cumulative effect of assisted reproduction procedures on placental development and epigenetic perturbations in a mouse model, Hum Mol Genet, vol.24, issue.24, pp.6975-6985, 2015.

L. Delle-piane, W. Lin, X. Liu, A. Donjacour, P. Minasi et al., Effect of the method of conception and embryo transfer procedure on mid-gestation placenta and fetal development in an IVF mouse model, Hum Reprod, vol.25, issue.8, pp.2039-2046, 2010.

M. M. Denomme and M. R. Mann, Genomic imprints as a model for the analysis of epigenetic stability during assisted reproductive technologies, Reproduction, vol.144, issue.4, pp.393-409, 2012.

L. Desch, C. Bruno, M. Luu, J. Barberet, C. Choux et al., Embryo multinucleation at the twocell stage is an independent predictor of intracytoplasmic sperm injection outcomes, Fertil Steril, vol.107, issue.1, pp.97-103, 2017.

A. I. Diplas, L. Lambertini, M. J. Lee, R. Sperling, Y. L. Lee et al., Differential expression of imprinted genes in normal and IUGR human placentas, Epigenetics, vol.4, issue.4, pp.235-240, 2009.

A. S. Doherty, M. R. Mann, K. D. Tremblay, B. Ms, and R. M. Schultz, Differential effects of culture on imprinted H19 expression in the preimplantation mouse embryo, Biol Reprod, vol.62, issue.6, pp.1526-1535, 2000.

D. C. Dolinoy, R. Das, J. R. Weidman, and R. L. Jirtle, Metastable epialleles, imprinting, and the fetal origins of adult diseases, Pediatr Res, vol.61, issue.5, pp.30-37, 2007.

J. J. Eckert, R. Porter, A. J. Watkins, E. Burt, S. Brooks et al., Metabolic induction and early responses of mouse blastocyst developmental programming following maternal low protein diet affecting life-long health, PLoS One, vol.7, issue.12, p.52791, 2012.

A. C. Eddy, H. Chapman, and E. M. George, Acute Hypoxia and Chronic Ischemia Induce Differential Total Changes in Placental Epigenetic Modifications, Reprod Sci, 2018.

N. El-hajj, L. Haertle, M. Dittrich, S. Denk, H. Lehnen et al., DNA methylation signatures in cord blood of ICSI children, Hum Reprod, vol.32, issue.8, pp.1761-1769, 2017.

J. G. Eriksson, E. Kajantie, K. L. Thornburg, O. C. Barker, and D. J. , Mother's body size and placental size predict coronary heart disease in men, Eur Heart J, vol.32, issue.18, pp.2297-2303, 2011.

A. Eskild, P. R. Romundstad, and L. J. Vatten, Placental weight and birthweight: does the association differ between pregnancies with and without preeclampsia?, Am J Obstet Gynecol, vol.201, issue.6, pp.595-591, 2009.

A. Farhi, B. Reichman, V. Boyko, A. Hourvitz, R. Lerner-geva et al., Maternal and neonatal health outcomes following assisted reproduction, Reprod Biomed Online, vol.26, issue.5, pp.454-461, 2013.

P. Fauque, Assistance Médicale à la Procréation: Conséquences épigénétiques et transcriptionnelles dans le modèle murin, 2009.

P. Fauque and . Bourc'his-d, Genes are not the whole story: retrotransposons as new determinants of male fertility. Textbook of Human Reproductive Genetics, pp.83-96, 2014.

P. Fauque, P. Jouannet, C. Lesaffre, M. A. Ripoche, L. Dandolo et al., Assisted Reproductive Technology affects developmental kinetics, H19 Imprinting Control Region methylation and H19 gene expression in individual mouse embryos, BMC Dev Biol, vol.7, p.116, 2007.

P. Fauque, M. A. Ripoche, J. Tost, L. Journot, A. Gabory et al., Modulation of imprinted gene network in placenta results in normal development of in vitro manipulated mouse embryos, Hum Mol Genet, vol.19, issue.9, pp.1779-1790, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01788884

D. Feil, M. Lane, C. T. Roberts, R. L. Kelley, L. J. Edwards et al., Effect of culturing mouse embryos under different oxygen concentrations on subsequent fetal and placental development, J Physiol, vol.572, pp.87-96, 2006.

A. L. Fortier, F. L. Lopes, N. Darricarrere, J. Martel, and J. M. Trasler, Superovulation alters the expression of imprinted genes in the midgestation mouse placenta, Hum Mol Genet, vol.17, issue.11, pp.1653-1665, 2008.

A. L. Fortier, S. Mcgraw, F. L. Lopes, K. M. Niles, M. Landry et al., Modulation of imprinted gene expression following superovulation, Mol Cell Endocrinol, vol.388, issue.1-2, pp.51-57, 2014.

A. L. Fowden, P. M. Coan, E. Angiolini, G. J. Burton, and C. M. , Imprinted genes and the epigenetic regulation of placental phenotype, Prog Biophys Mol Biol, vol.106, issue.1, pp.281-288, 2011.

M. Fujii, R. Matsuoka, E. Bergel, S. Van-der-poel, and T. Okai, Perinatal risk in singleton pregnancies after in vitro fertilization, Fertil Steril, vol.94, issue.6, pp.2113-2117, 2010.

A. Gabory, M. A. Ripoche, L. Digarcher, A. Watrin, F. Ziyyat et al., H19 acts as a trans regulator of the imprinted gene network controlling growth in mice, Development, vol.136, issue.20, pp.3413-3421, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01788895

J. Ghosh, C. Coutifaris, C. Sapienza, and M. Mainigi, Global DNA methylation levels are altered by modifiable clinical manipulations in assisted reproductive technologies, Clin Epigenetics, vol.9, p.14, 2017.

M. W. Gillman, Developmental origins of health and disease, N Engl J Med, vol.353, issue.17, pp.1848-1850, 2005.

P. Ginod, C. Choux, J. Barberet, T. Rousseau, C. Bruno et al., Singleton fetal growth kinetics depend on the mode of conception, Fertil Steril, vol.110, issue.6, pp.1109-1117, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01921821

C. Giorgetti, V. Meerschaut, F. , D. Roo, C. Saunier et al., Multivariate analysis identifies the estradiol level at ovulation triggering as an independent predictor of the first trimester pregnancy-associated plasma protein-A level in IVF/ICSI pregnancies, Hum Reprod, vol.28, issue.10, pp.2636-2642, 2013.

G. Giritharan, S. Talbi, A. Donjacour, D. Sebastiano, F. Dobson et al., Effect of in vitro fertilization on gene expression and development of mouse preimplantation embryos, Reproduction, vol.134, issue.1, pp.63-72, 2007.

A. C. Gjerris, A. Loft, A. Pinborg, M. Christiansen, and A. Tabor, First-trimester screening markers are altered in pregnancies conceived after IVF/ICSI, Ultrasound Obstet Gynecol, vol.33, issue.1, pp.8-17, 2009.

P. D. Gluckman, M. A. Hanson, C. C. Thornburg, and K. L. , Effect of in utero and early-life conditions on adult health and disease, N Engl J Med, vol.359, issue.1, pp.61-73, 2008.

M. G. Goll and T. H. Bestor, Eukaryotic cytosine methyltransferases, Annu Rev Biochem, vol.74, pp.481-514, 2005.

M. V. Gomes, J. Huber, R. A. Ferriani, A. Neto, A. M. Ramos et al., Abnormal methylation at the KvDMR1 imprinting control region in clinically normal children conceived by assisted reproductive technologies, Mol Hum Reprod, vol.15, issue.8, pp.471-477, 2009.

A. T. Grazul-bilska, M. L. Johnson, P. P. Borowicz, L. Baranko, R. Da et al., Placental development during early pregnancy in sheep: effects of embryo origin on fetal and placental growth and global methylation, Theriogenology, vol.79, issue.1, pp.94-102, 2013.

C. Haavaldsen, S. O. Samuelsen, and A. Eskild, The association of maternal age with placental weight: a population-based study of 536,954 pregnancies, BJOG, vol.118, issue.12, pp.1470-1476, 2011.

C. Haavaldsen, T. Tanbo, and A. Eskild, Placental weight in singleton pregnancies with and without assisted reproductive technology: a population study of 536,567 pregnancies, Hum Reprod, vol.27, issue.2, pp.576-582, 2012.

B. Haddad, F. Abirached, C. Louis-sylvestre, L. Blond, J. et al., Predictive value of early human chorionic gonadotrophin serum profiles for fetal growth retardation, Hum Reprod, vol.14, issue.11, pp.2872-2875, 1999.

R. Hart and R. J. Norman, The longer-term health outcomes for children born as a result of IVF treatment: Part I--General health outcomes, Hum Reprod Update, vol.19, issue.3, pp.232-243, 2013.

M. Hayashi, A. Nakai, S. Satoh, and Y. Matsuda, Adverse obstetric and perinatal outcomes of singleton pregnancies may be related to maternal factors associated with infertility rather than the type of assisted reproductive technology procedure used, Fertil Steril, vol.98, issue.4, pp.922-928, 2012.

D. L. Healy, S. Breheny, J. Halliday, A. Jaques, D. Rushford et al., Prevalence and risk factors for obstetric haemorrhage in 6730 singleton births after assisted reproductive technology in Victoria Australia, Hum Reprod, vol.25, issue.1, pp.265-274, 2010.

S. Hernandez-diaz, M. M. Werler, and A. A. Mitchell, Gestational hypertension in pregnancies supported by infertility treatments: role of infertility, treatments, and multiple gestations, Fertil Steril, vol.88, issue.2, pp.438-445, 2007.

S. Hiendleder, C. Mund, H. D. Reichenbach, H. Wenigerkind, G. Brem et al., Tissuespecific elevated genomic cytosine methylation levels are associated with an overgrowth phenotype of bovine fetuses derived by in vitro techniques, Biol Reprod, vol.71, issue.1, pp.217-223, 2004.

K. Hogg, E. M. Price, and W. P. Robinson, Improved reporting of DNA methylation data derived from studies of the human placenta, Epigenetics, vol.9, issue.3, pp.333-337, 2014.

N. Hori, M. Nagai, M. Hirayama, T. Hirai, K. Matsuda et al., Aberrant CpG methylation of the imprinting control region KvDMR1 detected in assisted reproductive technology-produced calves and pathogenesis of large offspring syndrome, Anim Reprod Sci, vol.122, issue.3-4, pp.303-312, 2010.

S. Houshdaran, C. R. Nezhat, K. C. Vo, Z. Zelenko, J. C. Irwin et al., Aberrant Endometrial DNA Methylome and Associated Gene Expression in Women with Endometriosis, Biol Reprod, vol.95, issue.5, p.93, 2016.

K. Y. Hsiao, M. H. Wu, and S. J. Tsai, Epigenetic regulation of the pathological process in endometriosis, Reprod Med Biol, vol.16, issue.4, pp.314-319, 2017.

X. L. Hu, C. Feng, X. H. Lin, Z. X. Zhong, Y. M. Zhu et al., High maternal serum estradiol environment in the first trimester is associated with the increased risk of small-for-gestational-age birth, J Clin Endocrinol Metab, vol.99, issue.6, pp.2217-2224, 2014.

Q. Huang, J. Li, F. Wang, M. T. Oliver, T. Tipton et al., Syncytin-1 modulates placental trophoblast cell proliferation by promoting G1/S transition, Cell Signal, vol.25, issue.4, pp.1027-1035, 2013.

S. Ibala-romdhane, A. , M. Khoueiry, R. Blachere, T. Guerin et al., Analysis of H19 methylation in control and abnormal human embryos, sperm and oocytes, Eur J Hum Genet, vol.19, issue.11, pp.1138-1143, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00646915

I. Iglesias-platas, A. Martin-trujillo, P. Petazzi, A. Guillaumet-adkins, M. Esteller et al., Altered expression of the imprinted transcription factor PLAGL1 deregulates a network of genes in the human IUGR placenta, Hum Mol Genet, vol.23, issue.23, pp.6275-6285, 2014.

R. S. Illingworth and A. P. Bird, CpG islands--'a rough guide, FEBS Lett, vol.583, issue.11, pp.1713-1720, 2009.

A. N. Imudia, A. O. Awonuga, J. O. Doyle, A. J. Kaimal, D. L. Wright et al., Peak serum estradiol level during controlled ovarian hyperstimulation is associated with increased risk of small for gestational age and preeclampsia in singleton pregnancies after in vitro fertilization, Fertil Steril, vol.97, issue.6, pp.1374-1379, 2012.

A. N. Imudia, R. H. Goldman, A. O. Awonuga, D. L. Wright, A. K. Styer et al., The impact of supraphysiologic serum estradiol levels on peri-implantation embryo development and early pregnancy outcome following in vitro fertilization cycles, J Assist Reprod Genet, vol.31, issue.1, pp.65-71, 2014.

O. Ishihara, R. Araki, A. Kuwahara, A. Itakura, H. Saito et al., Impact of frozen-thawed singleblastocyst transfer on maternal and neonatal outcome: an analysis of 277,042 single-embryo transfer cycles from 2008 to 2010 in Japan, Fertil Steril, vol.101, issue.1, pp.128-133, 2014.

R. A. Jackson, K. A. Gibson, Y. W. Wu, and M. S. Croughan, Perinatal outcomes in singletons following in vitro fertilization: a meta-analysis, Obstet Gynecol, vol.103, issue.3, pp.551-563, 2004.

M. Jahangiri, M. Shahhoseini, and B. Movaghar, H19 and MEST gene expression and histone modification in blastocysts cultured from vitrified and fresh two-cell mouse embryos, Reprod Biomed Online, vol.29, issue.5, pp.559-566, 2014.

P. A. Jones, Functions of DNA methylation: islands, start sites, gene bodies and beyond, Nat Rev Genet, vol.13, issue.7, pp.484-492, 2012.

B. Kallen, O. Finnstrom, K. G. Nygren, O. Olausson, P. Wennerholm et al., In vitro fertilisation in Sweden: obstetric characteristics, maternal morbidity and mortality, BJOG, vol.112, issue.11, pp.1529-1535, 2005.

D. Kanber, K. Buiting, M. Zeschnigk, L. M. Horsthemke, and B. , Low frequency of imprinting defects in ICSI children born small for gestational age, Eur J Hum Genet, vol.17, issue.1, pp.22-29, 2009.

H. Kano, I. Godoy, C. Courtney, M. R. Vetter, G. L. Gerton et al., L1 retrotransposition occurs mainly in embryogenesis and creates somatic mosaicism, Genes Dev, vol.23, issue.11, pp.1303-1312, 2009.

A. Katalinic, C. Rosch, and M. Ludwig, Pregnancy course and outcome after intracytoplasmic sperm injection: a controlled, prospective cohort study, Fertil Steril, vol.81, issue.6, pp.1604-1616, 2004.

S. Katari, N. Turan, M. Bibikova, O. Erinle, R. Chalian et al., DNA methylation and gene expression differences in children conceived in vitro or in vivo, Hum Mol Genet, vol.18, issue.20, pp.3769-3778, 2009.

S. Khosla, W. Dean, D. Brown, W. Reik, and R. Feil, Culture of preimplantation mouse embryos affects fetal development and the expression of imprinted genes, Biol Reprod, vol.64, issue.3, pp.918-926, 2001.

H. R. Kohan-ghadr, L. Kadam, C. Jain, A. Dr, and S. Drewlo, Potential role of epigenetic mechanisms in regulation of trophoblast differentiation, migration, and invasion in the human placenta, Cell Adh Migr, vol.10, issue.1-2, pp.126-135, 2016.

S. Korosec, H. Ban-frangez, I. Verdenik, U. Kladnik, V. Kotar et al., Singleton pregnancy outcomes after in vitro fertilization with fresh or frozen-thawed embryo transfer and incidence of placenta praevia, Biomed Res Int, p.431797, 2014.

J. Koudstaal, D. D. Braat, H. W. Bruinse, N. Naaktgeboren, J. P. Vermeiden et al., Obstetric outcome of singleton pregnancies after IVF: a matched control study in four Dutch university hospitals, Hum Reprod, vol.15, issue.8, pp.1819-1825, 2000.

O. Koukoura, S. Sifakis, G. Soufla, A. Zaravinos, S. Apostolidou et al., Loss of imprinting and aberrant methylation of IGF2 in placentas from pregnancies complicated with fetal growth restriction, Int J Mol Med, vol.28, issue.4, pp.481-487, 2011.

T. Kouzarides, Chromatin modifications and their function, Cell, vol.128, issue.4, pp.693-705, 2007.

L. Lambertini, S. R. Saul, A. B. Copperman, S. S. Hammerstad, Z. Yi et al., Intrauterine Reprogramming of the Polycystic Ovary Syndrome: Evidence from a Pilot Study of Cord Blood Global Methylation Analysis, Front Endocrinol (Lausanne), vol.8, p.352, 2017.

G. Lazaraviciute, M. Kauser, S. Bhattacharya, and P. Haggarty, A systematic review and meta-analysis of DNA methylation levels and imprinting disorders in children conceived by IVF/ICSI compared with children conceived spontaneously, Hum Reprod Update, vol.20, issue.6, pp.840-852, 2014.

P. E. Levi-setti, F. Cirillo, A. Smeraldi, E. Morenghi, G. Mulazzani et al., No advantage of fresh blastocyst versus cleavage stage embryo transfer in women under the age of 39: a randomized controlled study, J Assist Reprod Genet, vol.35, issue.3, pp.457-465, 2018.

T. Li, T. H. Vu, G. A. Ulaner, E. Littman, J. Q. Ling et al., IVF results in de novo DNA methylation and histone methylation at an Igf2-H19 imprinting epigenetic switch, Mol Hum Reprod, vol.11, issue.9, pp.631-640, 2005.

X. W. Liang, X. S. Cui, S. C. Sun, Y. X. Jin, Y. T. Heo et al., Superovulation induces defective methylation in line-1 retrotransposon elements in blastocyst, Reprod Biol Endocrinol, vol.11, p.69, 2013.

J. F. Litzky, S. L. Boulet, N. Esfandiari, Y. Zhang, D. M. Kissin et al., Effect of frozen/thawed embryo transfer on birthweight, macrosomia, and low birthweight rates in US singleton infants, Am J Obstet Gynecol, vol.218, issue.4, p.410, 2018.

J. F. Litzky, M. A. Deyssenroth, T. M. Everson, D. A. Armstrong, L. Lambertini et al., Placental imprinting variation associated with assisted reproductive technologies and subfertility, Epigenetics, vol.2017, pp.1-9

S. Liu, Y. Kuang, Y. Wu, Y. Feng, Q. Lyu et al., High oestradiol concentration after ovarian stimulation is associated with lower maternal serum beta-HCG concentration and neonatal birth weight, Reprod Biomed Online, vol.35, issue.2, pp.189-196, 2017.

S. P. Makaroun and K. P. Himes, Differential Methylation of Syncytin-1 and 2 Distinguishes Fetal Growth Restriction from Physiologic Small for Gestational Age, AJP Rep, vol.8, issue.1, pp.18-24, 2018.

S. S. Malchau, A. Loft, A. K. Henningsen, N. Andersen, A. Pinborg et al., Perinatal outcomes in 6,338 singletons born after intrauterine insemination in Denmark, Fertil Steril, vol.102, issue.4, pp.1110-1116, 2007.

E. Maman, E. Lunenfeld, A. Levy, H. Vardi, and G. Potashnik, Obstetric outcome of singleton pregnancies conceived by in vitro fertilization and ovulation induction compared with those conceived spontaneously, Fertil Steril, vol.70, issue.2, pp.240-245, 1998.

M. R. Mann, S. S. Lee, A. S. Doherty, R. I. Verona, L. D. Nolen et al., Selective loss of imprinting in the placenta following preimplantation development in culture, Development, vol.131, issue.15, pp.3727-3735, 2004.

B. A. Market-velker, A. D. Fernandes, and M. R. Mann, Side-by-side comparison of five commercial media systems in a mouse model: suboptimal in vitro culture interferes with imprint maintenance, Biol Reprod, vol.83, issue.6, pp.938-950, 2010.

B. A. Market-velker, L. Zhang, L. S. Magri, A. C. Bonvissuto, and M. R. Mann, Dual effects of superovulation: loss of maternal and paternal imprinted methylation in a dose-dependent manner, Hum Mol Genet, vol.19, issue.1, pp.36-51, 2010.

J. Mcminn, M. Wei, N. Schupf, J. Cusmai, E. B. Johnson et al., Unbalanced placental expression of imprinted genes in human intrauterine growth restriction, Placenta, vol.27, issue.6-7, pp.540-549, 2006.

N. Melamed, S. Choufani, L. E. Wilkins-haug, K. G. Weksberg, and R. , Comparison of genome-wide and gene-specific DNA methylation between ART and naturally conceived pregnancies, Epigenetics, vol.10, issue.6, pp.474-483, 2015.

C. Messerlian and A. J. Gaskins, Epidemiologic Approaches for Studying Assisted Reproductive Technologies: Design, Methods, Analysis and Interpretation, Curr Epidemiol Rep, vol.4, issue.2, pp.124-132, 2017.

D. M. Messerschmidt, Should I stay or should I go: protection and maintenance of DNA methylation at imprinted genes, Epigenetics, vol.7, issue.9, pp.969-975, 2012.

H. L. Miles, P. L. Hofman, J. Peek, M. Harris, D. Wilson et al., In vitro fertilization improves childhood growth and metabolism, J Clin Endocrinol Metab, vol.92, issue.9, pp.3441-3445, 2007.

J. R. Miles, C. E. Farin, K. F. Rodriguez, A. J. Farin, and P. W. , Angiogenesis and morphometry of bovine placentas in late gestation from embryos produced in vivo or in vitro, Biol Reprod, vol.71, issue.6, pp.1919-1926, 2004.

J. R. Miles, C. E. Farin, K. F. Rodriguez, A. J. Farin, and P. W. , Effects of embryo culture on angiogenesis and morphometry of bovine placentas during early gestation, Biol Reprod, vol.73, issue.4, pp.663-671, 2005.

R. E. Mills, E. A. Bennett, R. C. Iskow, and S. E. Devine, Which transposable elements are active in the human genome?, Trends Genet, vol.23, issue.4, pp.183-191, 2007.

D. Monk, P. Arnaud, S. Apostolidou, F. A. Hills, G. Kelsey et al., Limited evolutionary conservation of imprinting in the human placenta, Proc Natl Acad Sci, vol.103, issue.17, pp.6623-6628, 2006.
URL : https://hal.archives-ouvertes.fr/hal-01934548

M. Montagut, S. Santos-ribeiro, D. Vos, M. Polyzos, N. P. Drakopoulos et al., Frozenthawed embryo transfers in natural cycles with spontaneous or induced ovulation: the search for the best protocol continues, Hum Reprod, vol.31, issue.12, pp.2803-2810, 2016.

G. E. Moore, M. Ishida, C. Demetriou, L. Al-olabi, L. J. Leon et al., High FSH dosing is associated with reduced live birth rate in fresh but not subsequent frozen embryo transfers, Philos Trans R Soc Lond B Biol Sci, vol.370, pp.1-8, 1663.

Y. Nakamura, C. Yaguchi, H. Itoh, R. Sakamoto, T. Kimura et al., Morphologic characteristics of the placental basal plate in in vitro fertilization pregnancies: a possible association with the amount of bleeding in delivery, Hum Pathol, vol.46, issue.8, pp.1171-1179, 2015.

S. Nayak, M. E. Ochalski, B. Fu, K. M. Wakim, T. J. Chu et al., Progesterone level at oocyte retrieval predicts in vitro fertilization success in a short-antagonist protocol: a prospective cohort study, Fertil Steril, vol.101, issue.3, pp.676-682, 2014.

E. C. Nelissen, J. C. Dumoulin, F. Busato, L. Ponger, L. M. Eijssen et al., Altered gene expression in human placentas after IVF/ICSI, Hum Reprod, vol.29, issue.12, pp.2821-2831, 2014.

E. C. Nelissen, J. C. Dumoulin, A. Daunay, J. L. Evers, T. J. Van-montfoort et al., Placentas from pregnancies conceived by IVF/ICSI have a reduced DNA methylation level at the H19 and MEST differentially methylated regions, Hum Reprod, vol.28, issue.4, pp.1117-1126, 2013.

E. C. Nelissen, A. P. Van-montfoort, J. C. Dumoulin, and J. L. Evers, Epigenetics and the placenta, Hum Reprod Update, vol.17, issue.3, pp.397-417, 2011.

B. Novakovic and R. Saffery, The ever growing complexity of placental epigenetics -role in adverse pregnancy outcomes and fetal programming, Placenta, vol.33, issue.12, pp.959-970, 2012.

R. Ochsenkuhn, T. Strowitzki, M. Gurtner, A. Strauss, A. Schulze et al., Pregnancy complications, obstetric risks, and neonatal outcome in singleton and twin pregnancies after GIFT and IVF, Arch Gynecol Obstet, vol.268, issue.4, pp.256-261, 2003.

F. Olivennes, P. Rufat, B. Andre, A. Pourade, Q. Mc et al., The increased risk of complication observed in singleton pregnancies resulting from in-vitro fertilization (IVF) does not seem to be related to the IVF method itself, Hum Reprod, vol.8, issue.8, pp.1297-1300, 1993.

V. F. Oliver, H. L. Miles, W. S. Cutfield, P. L. Hofman, J. L. Ludgate et al., Defects in imprinting and genome-wide DNA methylation are not common in the in vitro fertilization population, Fertil Steril, vol.97, issue.1, pp.147-153, 2012.

K. Ozgur, H. Bulut, M. Berkkanoglu, P. Humaidan, and K. Coetzee, Artificial cryopreserved embryo transfer cycle success depends on blastocyst developmental rate and progesterone timing, Reprod Biomed Online, vol.36, issue.3, pp.269-276, 2018.

G. Palermo, H. Joris, D. P. Van-steirteghem, and A. C. , Pregnancies after intracytoplasmic injection of single spermatozoon into an oocyte, Lancet, vol.340, issue.8810, pp.17-18, 1992.

A. Pinborg, A. Loft, A. Henningsen, A. K. Rasmussen, S. Andersen et al., Infant outcome of 957 singletons born after frozen embryo replacement: the Danish National Cohort Study, Fertil Steril, vol.94, issue.4, pp.1320-1327, 1995.

A. Pinborg, U. B. Wennerholm, L. B. Romundstad, A. Loft, K. Aittomaki et al., Why do singletons conceived after assisted reproduction technology have adverse perinatal outcome? Systematic review and meta-analysis, Hum Reprod Update, vol.19, issue.2, pp.87-104, 2013.

P. Poikkeus, M. Gissler, L. Unkila-kallio, C. Hyden-granskog, and A. Tiitinen, Obstetric and neonatal outcome after single embryo transfer, Hum Reprod, vol.22, issue.4, pp.1073-1079, 2007.

B. Portha, A. Fournier, M. D. Kioon, M. V. Movassat, and J. , Early environmental factors, alteration of epigenetic marks and metabolic disease susceptibility, Biochimie, vol.97, pp.1-15, 2014.

J. Presl, 1st successful implantation of blastocyst, cultivated from the oocyte of treated woman after fertilization by husband's spermatozoa in vitro

, Cesk Gynekol, vol.42, issue.4, pp.296-297, 1977.

G. E. Ptak, A. Toschi, P. Fidanza, A. Zacchini, F. Czernik et al., Post-implantation mortality of in vitro produced embryos is associated with DNA methyltransferase 1 dysfunction in sheep placenta, Hum Reprod, vol.28, issue.2, pp.298-305, 2013.

S. E. Puumala, H. H. Nelson, J. A. Ross, R. H. Nguyen, M. A. Damario et al., Similar DNA methylation levels in specific imprinting control regions in children conceived with and without assisted reproductive technology: a cross-sectional study, BMC Pediatr, vol.12, p.33, 2012.

J. Qin, X. Liu, X. Sheng, W. H. Gao, and S. , Assisted reproductive technology and the risk of pregnancyrelated complications and adverse pregnancy outcomes in singleton pregnancies: a metaanalysis of cohort studies, Fertil Steril, vol.105, issue.1, pp.73-85, 2016.

B. Rahat, A. Mahajan, R. Bagga, H. A. Kaur, and J. , Epigenetic modifications at DMRs of placental genes are subjected to variations in normal gestation, pathological conditions and folate supplementation, Sci Rep, vol.7, p.40774, 2017.

R. C. Rancourt, H. Hr, and K. B. Michels, Methylation levels at imprinting control regions are not altered with ovulation induction or in vitro fertilization in a birth cohort, Hum Reprod, vol.27, issue.7, pp.2208-2216, 2012.

W. Reik, D. W. Walter, and J. , Epigenetic reprogramming in mammalian development, Science, vol.293, issue.5532, pp.1089-1093, 2001.

B. E. Reubinoff, A. Samueloff, M. Ben-haim, S. Friedler, J. G. Schenker et al., Is the obstetric outcome of in vitro fertilized singleton gestations different from natural ones? A controlled study, Fertil Steril, vol.67, issue.6, pp.1077-1083, 1997.

R. M. Rivera, P. Stein, J. R. Weaver, J. Mager, R. M. Schultz et al., Manipulations of mouse embryos prior to implantation result in aberrant expression of imprinted genes on day 9.5 of development, Hum Mol Genet, vol.17, issue.1, pp.1-14, 2008.

L. B. Romundstad, P. R. Romundstad, A. Sunde, V. During, V. Skjaerven et al., Increased risk of placenta previa in pregnancies following IVF/ICSI; a comparison of ART and non-ART pregnancies in the same mother, Hum Reprod, vol.21, issue.9, pp.2353-2358, 2006.

M. Roque, M. Valle, M. Sampaio, and S. Geber, Obstetric outcomes after fresh versus frozen-thawed embryo transfers: A systematic review and meta-analysis, JBRA Assist Reprod, vol.22, issue.3, pp.253-260, 2018.

T. Rousseau, C. Ferdynus, C. Quantin, G. Jb, and P. Sagot, Liveborn birth-weight of single and uncomplicated pregnancies between 28 and 42 weeks of gestation from Burgundy perinatal network

, J Gynecol Obstet Biol Reprod (Paris), vol.37, issue.6, pp.589-596, 2008.

M. Ruebner, P. L. Strissel, A. B. Ekici, E. Stiegler, U. Dammer et al., Reduced syncytin-1 expression levels in placental syndromes correlates with epigenetic hypermethylation of the ERVW-1 promoter region, PLoS One, vol.8, issue.2, p.56145, 2013.

M. Ruebner, P. L. Strissel, M. Langbein, F. Fahlbusch, D. L. Wachter et al., Impaired cell fusion and differentiation in placentae from patients with intrauterine growth restriction correlate with reduced levels of HERV envelope genes, J Mol Med (Berl), vol.88, issue.11, pp.1143-1156, 2010.

S. Sakian, K. Louie, E. C. Wong, J. Havelock, S. Kashyap et al., Altered gene expression of H19 and IGF2 in placentas from ART pregnancies, Placenta, vol.36, issue.10, pp.1100-1105, 2015.

S. D. Sakka, D. Loutradis, C. Kanaka-gantenbein, A. Margeli, M. Papastamataki et al., Absence of insulin resistance and low-grade inflammation despite early metabolic syndrome manifestations in children born after in vitro fertilization, Fertil Steril, vol.94, issue.5, pp.1693-1699, 2010.

F. Santos, L. Hyslop, P. Stojkovic, C. Leary, A. Murdoch et al., Evaluation of epigenetic marks in human embryos derived from IVF and ICSI, Hum Reprod, vol.25, issue.9, pp.2387-2395, 2010.

A. Sazonova, K. Kallen, A. Thurin-kjellberg, U. B. Wennerholm, and C. Bergh, Obstetric outcome in singletons after in vitro fertilization with cryopreserved/thawed embryos, Hum Reprod, vol.27, issue.5, pp.1343-1350, 2012.

S. Senapati, F. Wang, T. Ord, C. Coutifaris, R. Feng et al., Superovulation alters the expression of endometrial genes critical to tissue remodeling and placentation, J Assist Reprod Genet, vol.35, issue.10, pp.1799-1808, 2018.

T. Shevell, F. D. Malone, J. Vidaver, T. F. Porter, D. A. Luthy et al., Assisted reproductive technology and pregnancy outcome, Obstet Gynecol, vol.106, issue.5, pp.1039-1045, 2005.

X. Shi, Y. Ni, H. Zheng, S. Chen, M. Zhong et al., Abnormal methylation patterns at the IGF2/H19 imprinting control region in phenotypically normal babies conceived by assisted reproductive technologies, Eur J Obstet Gynecol Reprod Biol, vol.158, issue.1, pp.52-55, 2011.

R. A. Simmons, Developmental origins of adult disease, Pediatr Clin North Am, vol.56, issue.3, pp.449-466, 2009.

R. A. Simmons, T. Lj, and S. J. Gertz, Intrauterine growth retardation leads to the development of type 2 diabetes in the rat, Diabetes, vol.50, issue.10, pp.2279-2286, 2001.

K. D. Sinclair, L. E. Young, W. I. Mcevoy, and T. G. , In-utero overgrowth in ruminants following embryo culture: lessons from mice and a warning to men, Hum Reprod, vol.15, issue.5, pp.68-86, 2000.

S. Song, J. Ghosh, M. Mainigi, N. Turan, R. Weinerman et al., DNA methylation differences between in vitro-and in vivo-conceived children are associated with ART procedures rather than infertility, Clin Epigenetics, vol.7, p.41, 2015.

C. Sun, M. A. Velazquez, S. Marfy-smith, B. Sheth, A. Cox et al., Mouse early extraembryonic lineages activate compensatory endocytosis in response to poor maternal nutrition, Development, vol.141, issue.5, pp.1140-1150, 2014.

L. M. Sun, M. C. Walker, H. L. Cao, Q. Yang, T. Duan et al., Assisted reproductive technology and placenta-mediated adverse pregnancy outcomes, Obstet Gynecol, vol.114, issue.4, pp.818-824, 2009.

D. Tabs, T. Vejnovic, and N. Radunovic, Preeclampsia and eclampsia in parturients from the in vitro fertilization program, Med Pregl, vol.57, issue.1-2, pp.7-12, 2004.

C. P. Tallo, B. Vohr, W. Oh, L. P. Rubin, S. Db et al., Maternal and neonatal morbidity associated with in vitro fertilization, J Pediatr, vol.127, issue.5, pp.794-800, 1995.

K. Tan, Z. Zhang, K. Miao, Y. Yu, L. Sui et al., Dynamic integrated analysis of DNA methylation and gene expression profiles in in vivo and in vitro fertilized mouse post-implantation extraembryonic and placental tissues, Mol Hum Reprod, vol.22, issue.7, pp.485-498, 2016.

M. Y. Tan, L. C. Poon, D. L. Rolnik, A. Syngelaki, C. De-paco-matallana et al., Prediction and prevention of small-for-gestational-age neonates: evidence from SPREE and ASPRE, Ultrasound Obstet Gynecol, vol.52, issue.1, pp.52-59, 2018.

T. Tatsumi, S. C. Jwa, A. Kuwahara, M. Irahara, T. Kubota et al., No increased risk of major congenital anomalies or adverse pregnancy or neonatal outcomes following letrozole use in assisted reproductive technology, Hum Reprod, vol.32, issue.1, pp.125-132, 2017.

C. Thomopoulos, G. Salamalekis, K. Kintis, I. Andrianopoulou, H. Michalopoulou et al., Risk of hypertensive disorders in pregnancy following assisted reproductive technology: overview and meta-analysis, J Clin Hypertens (Greenwich), vol.19, issue.2, pp.173-183, 2017.

C. Thomopoulos, C. Tsioufis, H. Michalopoulou, T. Makris, V. Papademetriou et al., Assisted reproductive technology and pregnancy-related hypertensive complications: a systematic review, J Hum Hypertens, vol.27, issue.3, pp.148-157, 2013.

J. M. Thompson, L. M. Irgens, R. Skjaerven, and S. Rasmussen, Placenta weight percentile curves for singleton deliveries, BJOG, vol.114, issue.6, pp.715-720, 2007.

S. Tierling, N. Y. Souren, J. Gries, C. Loporto, M. Groth et al., Assisted reproductive technologies do not enhance the variability of DNA methylation imprints in human, J Med Genet, vol.47, issue.6, pp.371-376, 2010.

M. Toshimitsu, T. Nagamatsu, T. Nagasaka, Y. Iwasawa-kawai, A. Komatsu et al., Increased risk of pregnancy-induced hypertension and operative delivery after conception induced by in vitro fertilization/intracytoplasmic sperm injection in women aged 40 years and older, Fertil Steril, vol.102, issue.4, pp.1065-1070, 2014.

N. Turan, S. Katari, L. F. Gerson, R. Chalian, M. W. Foster et al., Inter-and intra-individual variation in allele-specific DNA methylation and gene expression in children conceived using assisted reproductive technology, PLoS Genet, vol.6, issue.7, p.1001033, 2010.

M. Turgal, E. Aydin, and O. Ozyuncu, Effect of micronized progesterone on fetal-placental volume in first-trimester threatened abortion, J Clin Ultrasound, vol.45, issue.1, pp.14-19, 2017.

D. Umlauf, Y. Goto, R. Cao, F. Cerqueira, A. Wagschal et al., Imprinting along the Kcnq1 domain on mouse chromosome 7 involves repressive histone methylation and recruitment of Polycomb group complexes, Nat Genet, vol.36, issue.12, pp.1296-1300, 2004.
URL : https://hal.archives-ouvertes.fr/hal-02262427

D. Umlauf, Y. Goto, and R. Feil, Site-specific analysis of histone methylation and acetylation, Methods Mol Biol, vol.287, pp.99-120, 2004.
URL : https://hal.archives-ouvertes.fr/hal-02262390

J. A. Van-den-hurk, I. C. Meij, M. C. Seleme, H. Kano, K. Nikopoulos et al., L1 retrotransposition can occur early in human embryonic development, Hum Mol Genet, 2007.

A. I. Van-der and D. , Superovulation of female mice delays embryonic and fetal development, Hum Reprod, vol.16, issue.6, pp.1237-1243, 2001.

A. P. Van-montfoort, L. L. Hanssen, D. Sutter, P. Viville, S. Geraedts et al., Assisted reproduction treatment and epigenetic inheritance, Hum Reprod Update, vol.18, issue.2, pp.171-197, 2012.

V. Borght, M. Wyns, and C. , Fertility and infertility: Definition and epidemiology, Clin Biochem, 2018.

A. Vargas, S. Zhou, M. Ethier-chiasson, D. Flipo, J. Lafond et al., Syncytin proteins incorporated in placenta exosomes are important for cell uptake and show variation in abundance in serum exosomes from patients with preeclampsia, FASEB J, vol.28, issue.8, pp.3703-3719, 2014.

S. Varmuza and K. Miri, What does genetics tell us about imprinting and the placenta connection?, Cell Mol Life Sci, vol.72, issue.1, pp.51-72, 2015.

A. Varrault, C. Gueydan, A. Delalbre, A. Bellmann, S. Houssami et al., Zac1 regulates an imprinted gene network critically involved in the control of embryonic growth, Dev Cell, vol.11, issue.5, pp.711-722, 2006.
URL : https://hal.archives-ouvertes.fr/inserm-00158399

R. N. Vincent, L. D. Gooding, K. Louie, C. Wong, E. Ma et al., Altered DNA methylation and expression of PLAGL1 in cord blood from assisted reproductive technology pregnancies compared with natural conceptions, Fertil Steril, vol.106, issue.3, pp.739-748, 2016.

L. A. Vrooman and M. S. Bartolomei, Can assisted reproductive technologies cause adult-onset disease? Evidence from human and mouse, Reprod Toxicol, vol.68, pp.72-84, 2017.

J. X. Wang, R. J. Norman, and A. J. Wilcox, Incidence of spontaneous abortion among pregnancies produced by assisted reproductive technology, Hum Reprod, vol.19, issue.2, pp.272-277, 2004.

Z. Wang, L. Xu, and F. He, Embryo vitrification affects the methylation of the H19/Igf2 differentially methylated domain and the expression of H19 and Igf2, Fertil Steril, vol.93, issue.8, pp.2729-2733, 2010.

N. Watanabe, T. Fujiwara, T. Suzuki, S. C. Jwa, K. Taniguchi et al., Is in vitro fertilization associated with preeclampsia? A propensity score matched study, BMC Pregnancy Childbirth, vol.14, p.69, 2014.

R. Weinerman, T. Ord, M. S. Bartolomei, C. Coutifaris, and M. Mainigi, The superovulated environment, independent of embryo vitrification, results in low birthweight in a mouse model, Biol Reprod, vol.97, issue.1, pp.133-142, 2017.

U. B. Wennerholm, A. K. Henningsen, L. B. Romundstad, C. Bergh, A. Pinborg et al., Perinatal outcomes of children born after frozen-thawed embryo transfer: a Nordic cohort study from the CoNARTaS group, Hum Reprod, vol.28, issue.9, pp.2545-2553, 2013.

N. Whitelaw, S. Bhattacharya, G. Hoad, G. W. Horgan, H. M. Haggarty et al., Epigenetic status in the offspring of spontaneous and assisted conception, Hum Reprod, vol.29, issue.7, pp.1452-1458, 2014.

E. C. Wong, C. Hatakeyama, R. Wp, and S. Ma, DNA methylation at H19/IGF2 ICR1 in the placenta of pregnancies conceived by in vitro fertilization and intracytoplasmic sperm injection, Fertil Steril, vol.95, issue.8, pp.2524-2526, 2011.

Y. Xiong, J. Wang, L. Liu, X. Chen, H. Xu et al., Effects of high progesterone level on the day of human chorionic gonadotrophin administration in in vitro fertilization cycles on epigenetic modification of endometrium in the peri-implantation period, Fertil Steril, vol.108, issue.2, pp.269-276, 2017.

X. Yang, Y. Li, L. C. Zhang, and W. , Current overview of pregnancy complications and live-birth outcome of assisted reproductive technology in mainland China, Fertil Steril, vol.101, issue.2, pp.385-391, 2014.

J. Yao, L. Geng, R. Huang, W. Peng, X. Chen et al., Effect of vitrification on in vitro development and imprinted gene Grb10 in mouse embryos, Reproduction, vol.154, issue.3, pp.97-105, 2017.

L. E. Young, K. D. Sinclair, and I. Wilmut, Large offspring syndrome in cattle and sheep, Rev Reprod, vol.3, issue.3, pp.155-163, 1998.

U. Zechner, G. Pliushch, E. Schneider, E. Hajj, N. Tresch et al., Quantitative methylation analysis of developmentally important genes in human pregnancy losses after ART and spontaneous conception, Mol Hum Reprod, vol.16, issue.9, pp.704-713, 2010.

Y. Zhang, Y. Cui, Z. Zhou, J. Sha, L. Y. Liu et al., Altered global gene expressions of human placentae subjected to assisted reproductive technology treatments, Placenta, vol.31, issue.4, pp.251-258, 2010.

Y. Zhang, Y. L. Zhang, C. Feng, Y. T. Wu, A. X. Liu et al., Comparative proteomic analysis of human placenta derived from assisted reproductive technology, Proteomics, vol.8, issue.20, pp.4344-4356, 2008.

Y. Zhang, W. Zhao, Y. Jiang, R. Zhang, J. Wang et al., Ultrastructural study on human placentae from women subjected to assisted reproductive technology treatments, Biol Reprod, vol.85, issue.3, pp.635-642, 2011.

L. Zhu, Y. Zhang, Y. Liu, R. Zhang, Y. Wu et al., Maternal and Live-birth Outcomes of Pregnancies following Assisted Reproductive Technology: A, Retrospective Cohort Study. Sci Rep, vol.6, 2016.

S. Dyer, G. M. Chambers, J. De-mouzon, K. G. Nygren, F. Zegers-hochschild et al., International Committee for Monitoring Assisted Reproductive Technologies world report: assisted reproductive technology, Hum Reprod, vol.31, pp.1588-609, 2008.

C. , C. De-geyter, C. Kupka, M. S. De-mouzon, J. Erb et al., Assisted reproductive technology in Europe, 2012: results generated from European registers by ESHRE, European IVF-Monitoring Consortium (EIM) for the European Society of Human Reproduction and Embryology (ESHRE), vol.31, pp.1638-52, 2016.

M. Ceelen, M. M. Van-weissenbruch, J. Vermeiden, F. E. Van-leeuwen, D. De-waal et al., Growth and development of children born after in vitro fertilization, Fertil Steril, vol.90, pp.1662-73, 2008.

P. Fauque, Ovulation induction and epigenetic anomalies, Fertil Steril, vol.99, pp.616-639, 2013.

A. Tandberg, K. Klungsøyr, L. B. Romundstad, and R. Skjaerven, Pre-eclampsia and assisted reproductive technologies: consequences of advanced maternal age, interbirth intervals, new partner and smoking habits, BJOG, vol.122, pp.915-937, 2015.

F. Thomson, S. Shanbhag, A. Templeton, and S. Bhattacharya, Obstetric outcome in women with subfertility, BJOG, vol.112, pp.632-639, 2005.

F. M. Helmerhorst, D. Perquin, D. Donker, and M. Keirse, Perinatal outcome of singletons and twins after assisted conception: a systematic review of controlled studies, BMJ, vol.328, p.261, 2004.

R. A. Jackson, K. A. Gibson, Y. W. Wu, and M. S. Croughan, Perinatal outcomes in singletons following in vitro fertilization: a meta-analysis, Obstet Gynecol, vol.103, pp.551-63, 2004.

T. Perri, R. Chen, R. Yoeli, P. Merlob, R. Orvieto et al., Are singleton assisted reproductive technology pregnancies at risk of prematurity?, J Assist Reprod Genet, vol.18, pp.245-254, 2001.

A. Pinborg, U. B. Wennerholm, L. B. Romundstad, A. Loft, K. Aittomaki et al., Why do singletons conceived after assisted reproduction technology have adverse perinatal outcome? Systematic review and meta-analysis, Hum Reprod Update, vol.19, pp.87-104, 2013.

C. De-geyter, M. De-geyter, S. Steimann, H. Zhang, and W. Holzgreve, Comparative birth weights of singletons born after assisted reproduction and natural conception in previously infertile women, Hum Reprod, vol.21, pp.705-717, 2006.

L. A. Schieve, C. Ferre, H. B. Peterson, M. Macaluso, M. A. Reynolds et al., Perinatal outcome among singleton infants conceived through assisted reproductive technology in the United States, Obstet Gynecol, vol.103, pp.1144-53, 2004.

L. A. Schieve, S. F. Meikle, C. Ferre, H. B. Peterson, G. Jeng et al., Low and very low birth weight in infants conceived with use of assisted reproductive technology, N Engl J Med, vol.346, pp.731-738, 2002.

E. S. Draper, J. J. Kurinczuk, K. R. Abrams, and M. Clarke, Assessment of separate contributions to perinatal mortality of infertility history and treatment: a casecontrol analysis, Lancet, vol.353, pp.1746-1755, 1999.

R. Isaksson and A. Tiitinen, Obstetric outcome in patients with unexplained infertility: comparison of treatment-related and spontaneous pregnancies, Acta Obstet Gynecol Scand, vol.77, pp.849-53, 1998.

A. Pinborg, A. A. Henningsen, A. Loft, S. S. Malchau, J. Forman et al., Large baby syndrome in singletons born after frozen embryo transfer (FET): Is it due to maternal factors or the cryotechnique?, Hum Reprod, vol.29, pp.618-645, 2014.

A. Pinborg, A. Loft, A. Henningsen, A. Rasmussen, S. Andersen et al., Infant outcome of 957 singletons born after frozen embryo replacement: the Danish National Cohort Study, Fertil Steril, vol.94, pp.1320-1327, 1995.

D. Barker, The origins of the developmental origins theory, J Intern Med, vol.261, pp.412-419, 2007.

P. D. Gluckman, M. A. Hanson, C. Cooper, and K. L. Thornburg, Effect of in utero and early-life conditions on adult health and disease, N Engl J Med, vol.359, pp.61-73, 2008.

P. D. Gluckman and M. A. Hanson, The developmental origins of the metabolic syndrome, Trends Endocrinol Metab, vol.15, pp.183-190, 2004.

K. M. Godfrey, K. A. Lillycrop, G. C. Burdge, P. D. Gluckman, and M. A. Hanson, Epigenetic mechanisms and the mismatch concept of the developmental origins of health and disease, Pediatr Res, vol.61, issue.5, pp.5-10, 2007.

R. Bukowski, G. Smith, F. D. Malone, R. H. Ball, D. A. Nyberg et al., Human sexual size dimorphism in early pregnancy, Am J Epidemiol, vol.165, pp.1216-1224, 2007.

T. Y. Leung, D. S. Sahota, L. W. Chan, L. W. Law, T. Y. Fung et al., Prediction of birth weight by fetal crown-rump length and maternal serum levels of pregnancy-associated plasma protein-A in the first trimester, Ultrasound Obstet Gynecol, vol.31, pp.10-14, 2008.

L. J. Salomon, S. Hourrier, R. Fanchin, Y. Ville, and P. Rozenberg, Is first-trimester crown-rump length associated with birthweight?, BJOG, vol.118, pp.1223-1231, 2011.

G. C. Smith, M. F. Smith, M. B. Mcnay, and J. E. Fleming, First-trimester growth and the risk of low birth weight, N Engl J Med, vol.339, pp.1817-1839, 1998.

M. Thorsell, M. Kaijser, H. Almstr?-om, and E. Andolf, Expected day of delivery from ultrasound dating versus last menstrual period-obstetric outcome when dates mismatch, BJOG, vol.115, pp.585-594, 2008.

E. M. Van-uitert, N. Exalto, G. J. Burton, S. P. Willemsen, A. Koning et al., Human embryonic growth trajectories and associations with fetal growth and birthweight, Hum Reprod, vol.28, pp.1753-61, 2013.

. Mook-kanamori, . Do, E. Steegers, P. H. Eilers, H. Raat et al., Risk factors and outcomes associated with first-trimester fetal growth restriction, JAMA, vol.303, pp.527-561, 2010.

I. Papastefanou, A. P. Souka, A. Pilalis, M. Eleftheriades, V. Michalitsi et al., First trimester prediction of small-and large-for-gestation neonates by an integrated model incorporating ultrasound parameters, biochemical indices and maternal characteristics, Acta Obstet Gynecol Scand, vol.91, pp.104-115, 2012.

J. Pardo, Y. Peled, Y. Yogev, N. Melamed, and A. Ben-haroush, Association of crown-rump length at 11 to 14 weeks' gestation and risk of a large-forgestational-age neonate, J Ultrasound Med, vol.29, pp.1315-1324, 2010.

L. Desch, C. Bruno, M. Luu, J. Barberet, C. Choux et al., Embryo multinucleation at the two-cell stage is an independent predictor of intracytoplasmic sperm injection outcomes, Fertil Steril, vol.107, pp.97-103, 2017.

L. J. Salomon,

, J Gynecol Obstet Biol Reprod (Paris), vol.40, pp.726-759, 2011.

, ACOG releases technical bulletin on preterm labor, Am Fam Physician, vol.52, pp.2105-2111, 1995.

, Diagnostic criteria and classification of hyperglycaemia first detected in pregnancy: a World Health Organization guideline, Diabetes Res Clin Pract, vol.103, pp.341-63, 2014.

T. Rousseau, C. Ferdynus, C. Quantin, J. Gouyon, P. Sagot et al., Liveborn birth-weight of single and uncomplicated pregnancies between 28 and 42 weeks of gestation from Burgundy perinatal network

, J Gynecol Obstet Biol Reprod (Paris), vol.37, pp.589-96, 2008.

, American College of Obstetricians and Gynecologists. ACOG practice bulletin no. 134: fetal growth restriction, Obstet Gynecol, vol.121, pp.1122-1155, 2013.

C. Vayssi-ere, L. Sentilhes, A. Ego, C. Bernard, D. Cambourieu et al., Fetal growth restriction and intra-uterine growth restriction: guidelines for clinical practice from the French College of Gynaecologists and Obstetricians, Eur J Obstet Gynecol Reprod Biol, vol.193, pp.10-18, 2015.

A. Herman, R. Maymon, E. Dreazen, E. Caspi, I. Bukovsky et al., Nuchal translucency audit: a novel image-scoring method, Ultrasound Obstet Gynecol, vol.12, pp.398-403, 1998.

F. P. Hadlock, R. B. Harrist, R. S. Sharman, R. L. Deter, and S. K. Park, Estimation of fetal weight with the use of head, body, and femur measurements-a prospective study, Am J Obstet Gynecol, vol.151, pp.333-340, 1985.

A. T. Papageorghiou, S. H. Kennedy, L. J. Salomon, E. O. Ohuma, C. Ismail et al., International standards for early fetal size and pregnancy dating based on ultrasound measurement of crown-rump length in the first trimester of pregnancy, Ultrasound Obstet Gynecol, vol.44, pp.641-649, 2014.

A. Ego, C. Prunet, B. Blondel, M. Kaminski, F. Goffinet et al., Customized and noncustomized French intrauterine growth curves. II-Comparison with existing curves and benefits of customization

, J Gynecol Obstet Biol Reprod, vol.45, pp.165-76, 2016.

A. Ego, C. Prunet, E. Lebreton, B. Blondel, M. Kaminski et al.,

, J Gynecol Obstet Biol Reprod (Paris), vol.45, pp.155-64, 2016.

F. P. Hadlock, R. B. Harrist, and J. Martinez-poyer, In utero analysis of fetal growth: a sonographic weight standard, Radiology, vol.181, pp.129-162, 1991.

J. Gardosi, M. Mongelli, M. Wilcox, and A. Chang, An adjustable fetal weight standard, Ultrasound Obstet Gynecol, vol.6, pp.168-74, 1995.

A. Ben-haroush, N. Melamed, G. Oron, I. Meizner, B. Fisch et al., Early first-trimester crown-rump length measurements in male and female singleton fetuses in IVF pregnancies, Ultrasound Obstet Gynecol, vol.25, pp.2610-2612, 2012.

K. E. O'neill, M. Tuuli, A. O. Odibo, R. R. Odem, and A. Cooper, Sex-related growth differences are present but not enhanced in in vitro fertilization pregnancies, Fertil Steril, vol.101, pp.407-419, 2014.

R. Bukowski, G. Smith, F. D. Malone, R. H. Ball, D. A. Nyberg et al., Fetal growth in early pregnancy and risk of delivering low birth weight infant: prospective cohort study, BMJ, vol.334, p.836, 2007.

M. Dommergues, R. Bessis, R. Henrion, and C. ,

, Gynecol Obstet Fertil, vol.34, pp.1090-1095, 2006.

M. Albouy-llaty, O. Thiebaugeorges, V. Goua, G. Magnin, M. Schweitzer et al., Influence of fetal and parental factors on intrauterine growth measurements: results of the EDEN mother-child cohort, Ultrasound Obstet Gynecol, vol.38, pp.673-80, 2011.

C. Bottomley, A. Daemen, F. Mukri, A. T. Papageorghiou, E. Kirk et al., Assessing first trimester growth: the influence of ethnic background and maternal age, Hum Reprod, vol.24, pp.284-90, 2009.

F. Lei, D. Liu, Y. Shen, L. Zhang, S. Li et al., Study on the influence of pregnancy-induced hypertension on neonatal birth weight, J Investig Med, vol.66, pp.1008-1022, 2018.

M. Bystr?-om, A. Liu, A. E. Quinton, B. L. Champion, K. Mann et al., Gestational diabetes independently increases birth length and augments the effects of maternal BMI on birth weight: a retrospective cohort study, Front Pediatr, vol.2, p.112, 2014.

A. Vambergue and I. Fajardy, Consequences of gestational and pregestational diabetes on placental function and birth weight, World J Diabetes, vol.2, pp.196-203, 2011.

M. Makgoba, M. D. Savvidou, and P. J. Steer, The effect of maternal characteristics and gestational diabetes on birthweight, BJOG, vol.119, pp.1091-1098, 2012.

E. M. Van-uitert, N. Van-der-elst-otte, J. J. Wilbers, N. Exalto, S. P. Willemsen et al., Periconception maternal characteristics and embryonic growth trajectories: the Rotterdam Predict study, Hum Reprod, vol.28, pp.3188-96, 2013.

M. A. Wilcox, C. S. Newton, and I. R. Johnson, Paternal influences on birthweight, Acta Obstet Gynecol Scand, vol.74, pp.15-23, 1995.

F. Wu, Y. Hwu, R. Lee, S. Li, F. Sun et al., First trimester ultrasound estimation of gestational age in pregnancies conceived after in vitro fertilization, Eur J Obstet Gynecol Reprod Biol, vol.160, pp.151-156, 2012.

S. C. Eindhoven, E. M. Van-uitert, J. Laven, S. P. Willemsen, A. Koning et al., The influence of IVF/ICSI treatment on human embryonic growth trajectories, Hum Reprod, vol.29, pp.2628-2664, 2014.

G. Hankins and M. Speer, Defining the pathogenesis and pathophysiology of neonatal encephalopathy and cerebral palsy, Obstet Gynecol, vol.102, pp.628-664, 2003.

M. Gaudoin, R. Dobbie, A. Finlayson, J. Chalmers, I. T. Cameron et al., Ovulation induction/intrauterine insemination in infertile couples is associated with low-birth-weight infants, Am J Obstet Gynecol, vol.188, pp.611-617, 2003.

C. Choux, V. Carmignac, C. Bruno, P. Sagot, D. Vaiman et al., The placenta: phenotypic and epigenetic modifications induced by assisted reproductive technologies throughout pregnancy, Clin Epigenetics, vol.7, p.87, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01188079

C. Thomopoulos, C. Tsioufis, H. Michalopoulou, T. Makris, V. Papademetriou et al., Assisted reproductive technology and pregnancy-related hypertensive complications: a systematic review, J Hum Hypertens, vol.27, pp.148-57, 2013.

W. Mak, L. A. Kondapalli, C. G. Gordon, J. Dimattina, M. Payson et al., Natural cycle IVF reduces the risk of low birthweight infants compared with conventional stimulated IVF, Hum Reprod, vol.31, pp.789-94, 2016.

C. Bourgain and P. Devroey, The endometrium in stimulated cycles for IVF, Hum Reprod Update, vol.9, pp.515-537, 2003.

D. Haouzi, S. Assou, C. Dechanet, T. Anahory, H. Dechaud et al., Controlled ovarian hyperstimulation for in vitro fertilization alters endometrial receptivity in humans: protocol effects, Biol Reprod, vol.82, pp.679-86, 2010.

J. A. Horcajadas, A. Riesewijk, J. Polman, R. Van-os, A. Pellicer et al., Effect of controlled ovarian hyperstimulation in IVF on endometrial gene expression profiles, Mol Hum Reprod, vol.11, pp.195-205, 2005.

B. A. Kolb and R. J. Paulson, The luteal phase of cycles utilizing controlled ovarian hyperstimulation and the possible impact of this hyperstimulation on embryo implantation, Am J Obstet Gynecol, vol.176, pp.1267-1276, 1997.

G. Ertzeid, R. Storeng, and T. Lyberg, Treatment with gonadotropins impaired implantation and fetal development in mice, J Assist Reprod Genet, vol.10, pp.286-91, 1993.

G. Ertzeid and R. Storeng, The impact of ovarian stimulation on implantation and fetal development in mice, Hum Reprod, vol.16, pp.221-226, 2001.

M. W. Gillman, Developmental origins of health and disease, N Engl J Med, vol.353, pp.1848-50, 2005.

M. J. Lambers, E. Groeneveld, D. A. Hoozemans, R. Schats, R. Homburg et al., Lower incidence of hypertensive complications during pregnancy in patients treated with low-dose aspirin during in vitro fertilization and early pregnancy, Hum Reprod, vol.24, pp.2447-50, 2009.

E. Bujold, S. Roberge, Y. Lacasse, M. Bureau, F. Audibert et al., Prevention of preeclampsia and intrauterine growth restriction with aspirin started in early pregnancy: a meta-analysis, Obstet Gynecol, vol.116, issue.2, pp.402-416, 2010.

J. T. Henderson, E. P. Whitlock, E. O'connor, C. A. Senger, J. H. Thompson et al., Low-dose aspirin for prevention of morbidity and mortality from preeclampsia: a systematic evidence review for the U.S. Preventive Services Task Force, Ann Intern Med, vol.160, pp.695-703, 2014.

J. L. Zhu, O. Basso, C. Obel, C. Bille, and J. Olsen, Infertility, infertility treatment, and congenital malformations: Danish national birth cohort, BMJ, vol.333, p.679, 2006.

, Four groups: in vitro fertilization (IVF), intracytoplasmic sperm microinjection (ICSI), frozen-embryo transfer (FET), and intrauterine insemination (IUI). BW ¼ birth weight

, CRL ¼ crown-rump length

, EFW ¼ estimated fetal weight. *EFW was not available owing to missing data

, **previous adverse perinatal outcomes defined as preterm delivery, 1004.

, Distribution of live-born singleton birth weights according to the mode of conception. This graph shows the percentage of newborns by birth weight category

. Ginod, Fetal growth and mode of conception, 2018 ARTICLE: EARLY PREGNANCY, 1177.

. Nathanielsz-;-gluckman, , 2006.

B. Vrooman, , 2017.

. Haavaldsen, Additionally, in animal models, many placental modifications have been reported after ART throughout pregnancy, Thus, as placental and foetal growth are closely linked and given the increased higher placental weight after IVF, ICSI or other methods of ART, 2004.

D. Piane, , 2010.

. Wegrzyn, Because placental weight cannot be assessed during pregnancy in humans, 2005.

. Rizzo, Several authors have investigated the relationships between first-trimester PV and adverse obstetric outcomes. PV has been shown to be predictive of preeclampsia, 2008.

. Arakaki, strongly correlated with birth weight and placental weight at birth, 2014.

. Rifouna, Despite the data, few studies have investigated PV after in vitro fertilization (IVF), and their results were conflicting, 2014.

. Sundheimer, Indeed, some studies found no significant difference in PV between IVF and spontaneous pregnancies at 10 WG, p.12, 2014.

(. Rizzo, but without providing a full vision of first-trimester markers and their relationship with perinatal and obstetrical outcomes. Yet, some first-trimester REFERENCES, 2016.

D. J. Amor, J. X. Xu, J. L. Halliday, I. Francis, D. L. Healy et al., Pregnancies conceived using assisted reproductive technologies (ART) have low levels of pregnancy-associated plasma protein-A (PAPP-A) leading to a high rate of false-positive results in first trimester screening for Down syndrome, Hum Reprod, vol.24, pp.1330-1338, 2009.

E. Anckaert, J. Schiettecatte, E. Sleurs, P. Devroey, and J. Smitz, First trimester screening for Down's syndrome after assisted reproductive technology: non-male factor infertility is associated with elevated free beta-human chorionic gonadotropin levels at 10-14 weeks of gestation, Fertil Steril, vol.90, pp.1206-1210, 2008.

T. Arakaki, J. Hasegawa, M. Nakamura, S. Hamada, M. Muramoto et al., Prediction of early-and late-onset pregnancy-induced hypertension using placental volume on three-dimensional ultrasound and uterine artery Doppler, Ultrasound Obstet Gynecol, vol.45, pp.539-543, 2015.

B. Ata and E. Seli, A universal freeze all strategy: why it is not warranted, Curr Opin Obstet Gynecol, vol.29, pp.136-145, 2017.

M. M. Aziz, G. Guirguis, S. Maratto, C. Benito, and E. J. Forman, Is there an association between assisted reproductive technologies and time and complications of the third stage of labor?, Arch Gynecol Obstet, vol.293, pp.1193-1196, 2016.

D. J. Barker, The origins of the developmental origins theory, J Intern Med, vol.261, pp.412-417, 2007.

D. J. Barker, A. R. Bull, C. Osmond, and S. J. Simmonds, Fetal and placental size and risk of hypertension in adult life, BMJ, vol.301, pp.259-262, 1990.

E. Bloise, W. Lin, X. Liu, R. Simbulan, K. S. Kolahi et al., Impaired placental nutrient transport in mice generated by in vitro fertilization, Endocrinology, vol.153, pp.3457-3467, 2012.

P. Cavoretto, V. Giorgione, S. Cipriani, P. Vigano, M. Candiani et al., Nuchal translucency measurement, free beta-hCG and PAPP-A concentrations in IVF/ICSI pregnancies: systematic review and metaanalysis, Prenat Diagn, vol.37, pp.540-555, 2017.

S. T. Chelbi and D. Vaiman, Genetic and epigenetic factors contribute to the onset of preeclampsia, Mol Cell Endocrinol, vol.282, pp.120-129, 2008.

K. B. Cheong, K. Y. Leung, T. K. Li, H. Y. Chan, Y. P. Lee et al., Comparison of inter-and intraobserver agreement and reliability between three different types of placental volume measurement technique (XI VOCAL, VOCAL and multiplanar) and validity in the in-vitro setting, Ultrasound Obstet Gynecol, vol.36, pp.210-217, 2010.

C. Choux, V. Carmignac, C. Bruno, P. Sagot, D. Vaiman et al., The placenta: phenotypic and epigenetic modifications induced by Assisted Reproductive Technologies throughout pregnancy, Clin Epigenetics, vol.7, p.87, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01188079

S. J. Churchill, E. T. Wang, M. Akhlaghpour, E. H. Goldstein, D. Eschevarria et al., Fertil Steril, vol.107, pp.1341-1347

. Cngof, , 2010.

, 39, S139, pp.338-142

L. A. Cole, Biological functions of hCG and hCG-related molecules, Reprod Biol Endocrinol, vol.8, p.102, 2010.

K. P. Conrad and V. L. Baker, Corpus luteal contribution to maternal pregnancy physiology and outcomes in assisted reproductive technologies, Am J Physiol Regul Integr Comp Physiol, vol.304, pp.69-72, 2013.

A. L. David and E. Jauniaux, Ultrasound and endocrinological markers of first trimester placentation and subsequent fetal size, Placenta, vol.40, pp.29-33, 2016.

L. Delle-piane, W. Lin, X. Liu, A. Donjacour, P. Minasi et al., Effect of the method of conception and embryo transfer procedure on mid-gestation placenta and fetal development in an IVF mouse model, Hum Reprod, vol.25, pp.2039-2046, 2010.

M. M. Denomme and M. R. Mann, Genomic imprints as a model for the analysis of epigenetic stability during assisted reproductive technologies, Reproduction, vol.144, pp.393-409, 2012.

L. Desch, C. Bruno, M. Luu, J. Barberet, C. Choux et al., Embryo multinucleation at the two-cell stage is an independent predictor of intracytoplasmic sperm injection outcomes, Fertil Steril, vol.107, pp.97-103, 2017.

S. Dyer, G. M. Chambers, J. De-mouzon, K. G. Nygren, F. Zegers-hochschild et al., International Committee for Monitoring Assisted Reproductive Technologies world report: Assisted Reproductive Technology, Hum Reprod, vol.31, pp.1588-1609, 2008.

M. Effendi, S. Demers, Y. Giguere, J. C. Forest, N. Brassard et al., Association between first-trimester placental volume and birth weight, Placenta, vol.35, pp.99-102, 2014.

P. Fauque, F. Mondon, F. Letourneur, M. A. Ripoche, L. Journot et al., In vitro fertilization and embryo culture strongly impact the placental transcriptome in the mouse model, PLoS One, vol.5, p.9218, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01788865

P. Fauque, M. A. Ripoche, J. Tost, L. Journot, A. Gabory et al., Modulation of imprinted gene network in placenta results in normal development of in vitro manipulated mouse embryos, Hum Mol Genet, vol.19, pp.1779-1790, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01788884

T. Fournier, K. Handschuh, V. Tsatsaris, J. Guibourdenche, and D. Evain-brion, Role of nuclear receptors and their ligands in human trophoblast invasion, J Reprod Immunol, vol.77, pp.161-170, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00175343

M. W. Gillman, Developmental origins of health and disease, N Engl J Med, vol.353, pp.1848-1850, 2005.

C. Giorgetti, F. Vanden-meerschaut, C. De-roo, O. Saunier, E. Quarello et al., Multivariate analysis identifies the estradiol level at ovulation triggering as an independent predictor of the first trimester pregnancy-associated plasma protein-A level in IVF/ICSI pregnancies, Hum Reprod, vol.28, pp.2636-2642, 2013.

G. Giritharan, S. Talbi, A. Donjacour, F. Di-sebastiano, A. T. Dobson et al., Effect of in vitro fertilization on gene expression and development of mouse preimplantation embryos, Reproduction, vol.134, pp.63-72, 2007.

A. C. Gjerris, A. Loft, A. Pinborg, M. Christiansen, and A. Tabor, First-trimester screening markers are altered in pregnancies conceived after IVF/ICSI, Ultrasound Obstet Gynecol, vol.33, pp.8-17, 2009.

P. D. Gluckman, M. A. Hanson, C. Cooper, and K. L. Thornburg, Effect of in utero and early-life conditions on adult health and disease, N Engl J Med, vol.359, pp.61-73, 2008.

N. L. Gonzalez-gonzalez, E. Gonzalez-davila, L. Gonzalez-marrero, E. Padron, J. R. Conde et al., Value of placental volume and vascular flow indices as predictors of intrauterine growth retardation, Eur J Obstet Gynecol Reprod Biol, vol.212, pp.13-19, 2017.

S. Gundu, M. Kulkarni, S. Gupte, A. Gupte, M. Gambhir et al., Correlation of first-trimester serum levels of pregnancy-associated plasma protein A with small-for-gestational-age neonates and preterm births, Int J Gynaecol Obstet, 2016.

C. Haavaldsen, T. Tanbo, and A. Eskild, Placental weight in singleton pregnancies with and without assisted reproductive technology: a population study of 536,567 pregnancies, Hum Reprod, vol.27, pp.576-582, 2012.

R. Hackmon, C. Librach, R. Burwick, N. Rodrigues, D. Farine et al., Do Early Fetal Measurements and Nuchal Translucency Correlate With Term Birth Weight?, J Obstet Gynaecol Can, vol.39, pp.750-756, 2017.

N. Hashish, A. Hassan, A. El-semary, R. Gohar, and M. A. Youssef, Could 3D placental volume and perfusion indices measured at 11-14 weeks predict occurrence of preeclampsia in high-risk pregnant women?, J Matern Fetal Neonatal Med, vol.28, pp.1094-1098, 2015.

D. L. Healy, S. Breheny, J. Halliday, A. Jaques, D. Rushford et al., Prevalence and risk factors for obstetric haemorrhage in 6730 singleton births after assisted reproductive technology in Victoria Australia, Hum Reprod, vol.25, pp.265-274, 2010.

X. L. Hu, C. Feng, X. H. Lin, Z. X. Zhong, Y. M. Zhu et al., High maternal serum estradiol environment in the first trimester is associated with the increased risk of small-for-gestational-age birth, J Clin Endocrinol Metab, vol.99, pp.2217-2224, 2014.

P. W. Hui, Y. H. Lam, M. H. Tang, E. H. Ng, W. S. Yeung et al., Maternal serum pregnancy-associated plasma protein-A and free beta-human chorionic gonadotrophin in pregnancies conceived with fresh and frozen-thawed embryos from in vitro fertilization and intracytoplasmic sperm injection, Prenat Diagn, vol.25, pp.390-393, 2005.

P. W. Hui, M. H. Tang, Y. H. Lam, E. H. Ng, W. S. Yeung et al., Maternal serum hCG and alpha-fetoprotein levels in pregnancies conceived after IVF or ICSI with fresh and frozen-thawed embryos, Hum Reprod, vol.18, pp.572-575, 2003.

L. P. Hunt, A. M. Mcinerney-leo, S. Sinnott, B. Sutton, R. Cincotta et al., Low first-trimester PAPP-A in IVF (fresh and frozenthawed) pregnancies, likely due to a biological cause, J Assist Reprod Genet, 2017.

A. N. Imudia, A. O. Awonuga, J. O. Doyle, A. J. Kaimal, D. L. Wright et al., Peak serum estradiol level during controlled ovarian hyperstimulation is associated with increased risk of small for gestational age and preeclampsia in singleton pregnancies after in vitro fertilization, Fertil Steril, vol.97, pp.1374-1379, 2012.

A. N. Imudia, A. O. Awonuga, A. J. Kaimal, D. L. Wright, A. K. Styer et al., Elective cryopreservation of all embryos with subsequent cryothaw embryo transfer in patients at risk for ovarian hyperstimulation syndrome reduces the risk of adverse obstetric outcomes: a preliminary study, Fertil Steril, vol.99, pp.168-173, 2013.

A. R. Isles and A. J. Holland, Imprinted genes and mother-offspring interactions, Early Hum Dev, vol.81, pp.73-77, 2005.

R. A. Jackson, K. A. Gibson, Y. W. Wu, and M. S. Croughan, Perinatal outcomes in singletons following in vitro fertilization: a meta-analysis, Obstet Gynecol, vol.103, pp.551-563, 2004.

D. J. Kaser, A. Melamed, C. L. Bormann, D. E. Myers, S. A. Missmer et al., Cryopreserved embryo transfer is an independent risk factor for placenta accreta, Fertil Steril, vol.103, pp.1176-1184, 2015.

S. Liu, Y. Kuang, Y. Wu, Y. Feng, Q. Lyu et al., High oestradiol concentration after ovarian stimulation is associated with lower maternal serum beta-HCG concentration and neonatal birth weight, Reprod Biomed Online, vol.35, pp.189-196, 2017.

C. Messerlian and A. J. Gaskins, Epidemiologic Approaches for Studying Assisted Reproductive Technologies: Design, Methods, Analysis and Interpretation, Curr Epidemiol Rep, vol.4, pp.124-132, 2017.

J. R. Miles, C. E. Farin, K. F. Rodriguez, J. E. Alexander, and P. W. Farin, Angiogenesis and morphometry of bovine placentas in late gestation from embryos produced in vivo or in vitro, Biol Reprod, vol.71, pp.1919-1926, 2004.

J. R. Miles, C. E. Farin, K. F. Rodriguez, J. E. Alexander, and P. W. Farin, Effects of embryo culture on angiogenesis and morphometry of bovine placentas during early gestation, Biol Reprod, vol.73, pp.663-671, 2005.

M. Montagut, S. Santos-ribeiro, M. De-vos, N. P. Polyzos, P. Drakopoulos et al., Frozen-thawed embryo transfers in natural cycles with spontaneous or induced ovulation: the search for the best protocol continues, Hum Reprod, vol.31, pp.2803-2810, 2016.

G. Moser and B. Huppertz, Implantation and extravillous trophoblast invasion: From rare archival specimens to modern biobanking, Placenta, vol.56, pp.19-26, 2017.

E. M. Munch, A. E. Sparks, M. B. Zimmerman, B. J. Van-voorhis, and E. H. Duran, High FSH dosing is associated with reduced live birth rate in fresh but not subsequent frozen embryo transfers, Hum Reprod, pp.1-8, 2017.

Y. Nakamura, C. Yaguchi, H. Itoh, R. Sakamoto, T. Kimura et al., Morphologic characteristics of the placental basal plate in in vitro fertilization pregnancies: a possible association with the amount of bleeding in delivery, Hum Pathol, vol.46, pp.1171-1179, 2015.

P. W. Nathanielsz, Animal models that elucidate basic principles of the developmental origins of adult diseases, ILAR J, vol.47, pp.73-82, 2006.

F. Ouyang, M. G. Parker, Z. C. Luo, X. Wang, H. J. Zhang et al., Maternal BMI, gestational diabetes, and weight gain in relation to childhood obesity: The mediation effect of placental weight, Obesity (Silver Spring), vol.24, pp.938-946, 2016.

K. Ozgur, H. Bulut, M. Berkkanoglu, P. Humaidan, and K. Coetzee, Artificial cryopreserved embryo transfer cycle success depends on blastocyst developmental rate and progesterone timing, Reprod Biomed Online, vol.36, pp.269-276, 2018.

A. T. Papageorghiou, S. H. Kennedy, L. J. Salomon, E. O. Ohuma, L. Ismail et al., International standards for early fetal size and pregnancy dating based on ultrasound measurement of crown-rump length in the first trimester of pregnancy, Ultrasound Obstet Gynecol, vol.44, pp.641-648, 2014.

I. Papastefanou, C. Chrelias, C. Siristatidis, D. Kappou, M. Eleftheriades et al., Placental volume at 11 to 14 gestational weeks in pregnancies complicated with fetal growth restriction and preeclampsia, 2018.

A. Pinborg, A. A. Henningsen, A. Loft, S. S. Malchau, J. Forman et al., Large baby syndrome in singletons born after frozen embryo transfer (FET): is it due to maternal factors or the cryotechnique?, Hum Reprod, vol.29, pp.618-627, 2014.

F. Prefumo, N. Fratelli, S. C. Soares, and B. Thilaganathan, Uterine artery Doppler velocimetry at 11-14 weeks in singleton pregnancies conceived by assisted reproductive technology, Ultrasound Obstet Gynecol, vol.29, pp.141-145, 2007.

F. Prefumo, N. J. Sebire, and B. Thilaganathan, Decreased endovascular trophoblast invasion in first trimester pregnancies with high-resistance uterine artery Doppler indices, Hum Reprod, vol.19, pp.206-209, 2004.

G. E. Ptak, A. Toschi, P. Fidanza, A. Zacchini, F. Czernik et al., Post-implantation mortality of in vitro produced embryos is associated with DNA methyltransferase 1 dysfunction in sheep placenta, Hum Reprod, vol.28, pp.298-305, 2013.

L. P. Reynolds and J. S. Caton, Role of the pre-and post-natal environment in developmental programming of health and productivity, Mol Cell Endocrinol, vol.354, pp.54-59, 2012.

M. S. Rifouna, A. D. Reus, A. H. Koning, P. J. Van-der-spek, N. Exalto et al., First trimester trophoblast and placental bed vascular volume measurements in IVF or IVF/ICSI pregnancies, Hum Reprod, vol.29, pp.2644-2649, 2014.

G. Rizzo, E. Aiello, M. E. Pietrolucci, and D. Arduini, Placental volume and uterine artery doppler evaluation at 11 + 0 to 13 + 6 weeks of gestation in pregnancies conceived with in vitro fertilization: comparison between autologous and donor oocyte recipients, Ultrasound Obstet Gynecol, 2015.

G. Rizzo, E. Aiello, M. E. Pietrolucci, and D. Arduini, Are There Differences in Placental Volume and Uterine Artery Doppler in Pregnancies Resulting From the Transfer of Fresh Versus Frozen-Thawed Embryos Through In Vitro Fertilization, Reprod Sci, 2016.

G. Rizzo, A. Capponi, O. Cavicchioni, M. Vendola, and D. Arduini, First trimester uterine Doppler and three-dimensional ultrasound placental volume calculation in predicting pre-eclampsia, Eur J Obstet Gynecol Reprod Biol, vol.138, pp.147-151, 2008.

T. Rousseau, C. Ferdynus, C. Quantin, J. B. Gouyon, and P. Sagot, Liveborn birthweight of single and uncomplicated pregnancies between 28 and 42 weeks of gestation from Burgundy perinatal network, 2008.

, J Gynecol Obstet Biol Reprod (Paris), vol.37, pp.589-596

L. J. Salomon, , 2011.

, J Gynecol Obstet Biol Reprod (Paris), vol.40, pp.726-733

N. Schwartz, M. D. Sammel, R. Leite, and S. Parry, First-trimester placental ultrasound and maternal serum markers as predictors of small-for-gestational-age infants, Am J Obstet Gynecol, vol.211, pp.251-258, 2014.

L. Sentilhes, C. Vayssiere, C. Deneux-tharaux, A. G. Aya, F. Bayoumeu et al., Postpartum hemorrhage: guidelines for clinical practice from the French College of Gynaecologists and Obstetricians (CNGOF), 2016.
URL : https://hal.archives-ouvertes.fr/hal-01482637

, Eur J Obstet Gynecol Reprod Biol, vol.198, pp.12-21

P. Shwarzman, A. Y. Waintraub, M. Frieger, A. Bashiri, M. Mazor et al., Third-trimester abnormal uterine artery Doppler findings are associated with adverse pregnancy outcomes, J Ultrasound Med, vol.32, pp.2107-2113, 2013.

P. Sirikunalai, C. Wanapirak, S. Sirichotiyakul, F. Tongprasert, K. Srisupundit et al., Associations between maternal serum free beta human chorionic gonadotropin (beta-hCG) levels and adverse pregnancy outcomes, J Obstet Gynaecol, vol.36, pp.178-182, 2016.

S. Spijkers, J. W. Lens, R. Schats, and C. B. Lambalk, Fresh and Frozen-Thawed Embryo Transfer Compared to Natural Conception: Differences in Perinatal Outcome?, Gynecol Obstet Invest, 2017.

T. Stampalija, L. Monasta, D. D. Di-martino, M. Quadrifoglio, L. Lo-bello et al., The association of first trimester uterine arteries Doppler velocimetry with different clinical phenotypes of hypertensive disorders of pregnancy: a longitudinal study, J Matern Fetal Neonatal Med, pp.1-9, 2017.

L. W. Sundheimer, J. L. Chan, R. Buttle, R. Dipentino, O. Muramoto et al., Mode of conception does not affect fetal or placental growth parameters or ratios in early gestation or at delivery, J Assist Reprod Genet, 2018.

M. Y. Tan, L. C. Poon, D. L. Rolnik, A. Syngelaki, C. De-paco-matallana et al., Prediction and prevention of small-for-gestational-age neonates: evidence from SPREE and ASPRE, Ultrasound Obstet Gynecol, vol.52, pp.52-59, 2018.

M. Y. Tan, D. Wright, A. Syngelaki, R. Akolekar, S. Cicero et al., Comparison of diagnostic accuracy of early screening for pre-eclampsia by NICE guidelines and a method combining maternal factors and biomarkers: results of SPREE, Ultrasound Obstet Gynecol, vol.51, pp.743-750, 2018.

C. Thomopoulos, C. Tsioufis, H. Michalopoulou, T. Makris, V. Papademetriou et al., Assisted reproductive technology and pregnancy-related hypertensive complications: a systematic review, J Hum Hypertens, vol.27, pp.148-157, 2013.

N. Tul and Z. Novak-antolic, Serum PAPP-A levels at 10-14 weeks of gestation are altered in women after assisted conception, Prenat Diagn, vol.26, pp.1206-1211, 2006.

M. Turgal, E. Aydin, and O. Ozyuncu, Effect of micronized progesterone on fetalplacental volume in first-trimester threatened abortion, J Clin Ultrasound, vol.45, pp.14-19, 2017.

M. Vander-borght and C. Wyns, Fertility and infertility: Definition and epidemiology, Clin Biochem, 2018.

L. A. Vrooman and M. S. Bartolomei, Can assisted reproductive technologies cause adult-onset disease? Evidence from human and mouse, Reprod Toxicol, vol.68, pp.72-84, 2017.

P. Wegrzyn, C. Faro, O. Falcon, C. F. Peralta, and K. H. Nicolaides, Placental volume measured by three-dimensional ultrasound at 11 to 13 + 6 weeks of gestation: relation to chromosomal defects, Ultrasound Obstet Gynecol, vol.26, pp.28-32, 2005.

R. Weinerman and M. Mainigi, Why we should transfer frozen instead of fresh embryos: the translational rationale, Fertil Steril, vol.102, pp.10-18, 2014.

Y. Andersen, C. Vilbour-andersen, and K. , Improving the luteal phase after ovarian stimulation: reviewing new options, Reprod Biomed Online, vol.28, pp.552-559, 2014.

J. L. Yovich, J. L. Conceicao, J. D. Stanger, P. M. Hinchliffe, and K. N. Keane, Midluteal serum progesterone concentrations govern implantation rates for cryopreserved embryo transfers conducted under hormone replacement, Reprod Biomed Online, vol.31, pp.180-191, 2015.

B. Yucel, A. Gedikbasi, O. Dundar, Y. Olgac, D. Yildirim et al., The utility of first trimester uterine artery Doppler, placental volume and PAPP-A levels alone and in combination to predict preeclampsia, Pregnancy Hypertens, vol.6, pp.269-273, 2016.

S. G. Zhabin, V. S. Gorin, and N. S. Judin, Review: immunomodulatory activity of pregnancy-associated plasma protein-A, J Clin Lab Immunol, vol.52, pp.41-50, 2003.

Y. Zhang, Y. L. Zhang, C. Feng, Y. T. Wu, A. X. Liu et al., Comparative proteomic analysis of human placenta derived from assisted reproductive technology, Proteomics, vol.8, pp.4344-4356, 2008.

X. L. Zhu, J. Wang, R. Z. Jiang, and Y. C. Teng, Pulsatility index in combination with biomarkers or mean arterial pressure for the prediction of pre-eclampsia: Systematic literature review and meta-analysis, Ann Med, vol.47, pp.414-422, 2015.