J. Cha, X. Sun, and S. Dey, Mechanisms of implantation: strategies for successful pregnancy, Nature Medicine, vol.8, issue.12, pp.1754-1767, 2012.
DOI : 10.1016/j.devcel.2012.09.002

H. Cakmak and H. Taylor, Implantation failure: molecular mechanisms and clinical treatment, Human Reproduction Update, vol.17, issue.2, pp.242-253, 2011.
DOI : 10.1093/humupd/dmq037

N. Elaine and . Marieb, Biologie Humaine, Principes d'anatomie et de physiologie, 2008.

S. Storck, A. Health-medline-chapman, W. Abraham, S. Zamudio, C. Coffin et al., Ref Type: Internet Communication http://www.nlm.nih.gov/medlineplus/ency/patientinstructions/000603.htm 7 Temporal relationships between hormonal and hemodynamic changes in early human pregnancy, Kidney Int, vol.54, pp.23-31, 1998.

A. Malassine and L. Cronier, Hormones and Human Trophoblast Differentiation, Endocrine, vol.19, issue.1, pp.3-11, 2002.
DOI : 10.1385/ENDO:19:1:3

A. Enders and A. Carter, Review: The evolving placenta: Different developmental paths to a hemochorial relationship, Placenta, vol.33, pp.92-98, 2012.
DOI : 10.1016/j.placenta.2011.10.009

J. Frost and G. Moore, The Importance of Imprinting in the Human Placenta, PLoS Genetics, vol.6, issue.7, p.1001015, 2010.
DOI : 10.1371/journal.pgen.1001015.t002

D. Goldman-wohl and S. Yagel, Regulation of trophoblast invasion: from normal implantation to pre-eclampsia, Molecular and Cellular Endocrinology, vol.187, issue.1-2, pp.233-238, 2002.
DOI : 10.1016/S0303-7207(01)00687-6

P. Kaufmann, S. Black, and B. Huppertz, Endovascular Trophoblast Invasion: Implications for the Pathogenesis of Intrauterine Growth Retardation and Preeclampsia, Biology of Reproduction, vol.69, issue.1, pp.1-7, 2003.
DOI : 10.1095/biolreprod.102.014977

L. Rockwell, E. Vargas, and L. Moore, Human physiological adaptation to pregnancy: Inter- and intraspecific perspectives, American Journal of Human Biology, vol.79, issue.3, pp.330-341, 2003.
DOI : 10.1002/ajhb.10151

L. Lunghi, M. Ferretti, S. Medici, C. Biondi, and F. Vesce, Control of human trophoblast function, Reproductive Biology and Endocrinology, vol.5, issue.1, p.6, 2007.
DOI : 10.1186/1477-7827-5-6

S. Lager and T. Powell, Regulation of Nutrient Transport across the Placenta, Journal of Pregnancy, vol.62, issue.5, p.179827, 2012.
DOI : 10.1016/j.placenta.2009.12.009

A. Bell and R. Ehrhardt, Regulation of placental nutrient transport and implications for fetal growth, Nutrition Research Reviews, vol.190, issue.02, pp.211-230, 2002.
DOI : 10.1203/00006450-198603000-00015

M. Militello, E. Pappalardo, S. Ermito, A. Dinatale, A. Cavaliere et al., Obstetric management of IUGR, J Prenat Med, vol.3, pp.6-9, 2009.

B. Young, R. Levine, and S. Karumanchi, Pathogenesis of Preeclampsia, Annual Review of Pathology: Mechanisms of Disease, vol.5, issue.1, pp.173-192, 2010.
DOI : 10.1146/annurev-pathol-121808-102149

S. Srinivas, A. Edlow, P. Neff, M. Sammel, C. Andrela et al., Rethinking IUGR in preeclampsia: dependent or independent of maternal hypertension?, Journal of Perinatology, vol.111, issue.10, pp.680-684, 2009.
DOI : 10.1038/jp.2009.83

A. Mccarthy, R. Woolfson, S. Raju, and L. Poston, Abnormal endothelial cell function of resistance arteries from women with preeclampsia, American Journal of Obstetrics and Gynecology, vol.168, issue.4, pp.1323-1330, 1993.
DOI : 10.1016/0002-9378(93)90389-Z

C. Yallampalli, Y. Dong, and S. Wimalawansa, Calcitonin gene-related peptide reverses the hypertension and significantly decreases the fetal mortality in pre-eclampsia rats induced by NG-nitro-L-arginine methyl ester, Human Reproduction, vol.11, issue.4, pp.895-899, 1996.
DOI : 10.1093/oxfordjournals.humrep.a019274

A. Friel, D. Sexton, O. Reilly, M. Smith, T. Morrison et al., Rho A/Rho kinase: human umbilical artery mRNA expression in normal and pre eclamptic pregnancies and functional role in isoprostane-induced vasoconstriction, Reproduction, vol.132, issue.1, pp.169-176, 2006.
DOI : 10.1530/rep.1.01088

G. Amberg and M. Navedo, Calcium Dynamics in Vascular Smooth Muscle, Microcirculation, vol.298, issue.Pt 3, pp.281-289, 2013.
DOI : 10.1111/micc.12046

S. Tawara and H. Shimokawa, Progress of the Study of Rho-kinase and Future Perspective of the Inhibitor, YAKUGAKU ZASSHI, vol.127, issue.3, pp.501-514, 2007.
DOI : 10.1248/yakushi.127.501

P. Kafka, O. Vajnerova, J. Herget, and V. Hampl, Rho-kinase inhibition attenuates acute hypoxic fetoplacental vasoconstriction in the rat 1, Physiol Res, vol.61, issue.2, pp.43-48, 2012.

M. Olson, Applications for ROCK kinase inhibition, Current Opinion in Cell Biology, vol.20, issue.2, pp.242-248, 2008.
DOI : 10.1016/j.ceb.2008.01.002

S. Satoh, A. Hitomi, I. Ikegaki, K. Kawasaki, O. Nakazono et al., Amelioration of endothelial damage/dysfunction is a possible mechanism for the neuroprotective effects of Rho-kinase inhibitors against ischemic brain damage, Brain Research Bulletin, vol.81, issue.1, pp.191-195, 2010.
DOI : 10.1016/j.brainresbull.2009.08.021

H. Fujita, Y. Fukumoto, K. Saji, K. Sugimura, J. Demachi et al., Acute vasodilator effects of inhaled fasudil, a specific Rho-kinase inhibitor, in patients with pulmonary arterial hypertension, Heart and Vessels, vol.50, issue.Pt2, pp.144-149, 2010.
DOI : 10.1007/s00380-009-1176-8

T. Frayling, N. Timpson, M. Weedon, E. Zeggini, R. Freathy et al., A Common Variant in the FTO Gene Is Associated with Body Mass Index and Predisposes to Childhood and Adult Obesity, Science, vol.316, issue.5826, pp.889-894, 2007.
DOI : 10.1126/science.1141634

I. , K. Health, and R. , Enquête épidémiologique nationale sur le surpoids et l'obésité ObEpi-Roche, 2012.

L. Poston, Maternal obesity, gestational weight gain and diet as determinants of offspring long term health, Best Practice & Research Clinical Endocrinology & Metabolism, vol.26, issue.5, pp.627-639, 2012.
DOI : 10.1016/j.beem.2012.03.010

E. Buschur and C. Kim, Guidelines and interventions for obesity during pregnancy, International Journal of Gynecology & Obstetrics, vol.19, issue.(1), pp.6-10, 2012.
DOI : 10.1016/j.ijgo.2012.04.025

C. Olson, M. Strawderman, P. Hinton, and T. Pearson, Gestational weight gain and postpartum behaviors associated with weight change from early pregnancy to 1???y postpartum, International Journal of Obesity, vol.27, issue.1, pp.117-127, 2003.
DOI : 10.1038/sj.ijo.0802156

S. Phelan, Pregnancy: a ???teachable moment??? for weight control and obesity prevention, American Journal of Obstetrics and Gynecology, vol.202, issue.2, pp.135-138, 2010.
DOI : 10.1016/j.ajog.2009.06.008

A. Siega-riz, M. Viswanathan, M. Moos, A. Deierlein, S. Mumford et al., A systematic review of outcomes of maternal weight gain according to the Institute of Medicine recommendations: birthweight, fetal growth, and postpartum weight retention, American Journal of Obstetrics and Gynecology, vol.201, issue.4, pp.339-353, 2009.
DOI : 10.1016/j.ajog.2009.07.002

Y. Linne, L. Dye, B. Barkeling, and S. Rossner, Long-Term Weight Development in Women: A 15-Year Follow-up of the Effects of Pregnancy, Obesity Research, vol.123, issue.7, pp.1166-1178, 2004.
DOI : 10.1038/oby.2004.146

E. Davis, S. Zyzanski, C. Olson, K. Stange, and R. Horwitz, Racial, Ethnic, and Socioeconomic Differences in the Incidence of Obesity Related to Childbirth, American Journal of Public Health, vol.99, issue.2, pp.294-299, 2009.
DOI : 10.2105/AJPH.2007.132373

A. Frias and K. Grove, Obesity: a transgenerational problem linked to nutrition during pregnancy, Semin Reprod Med, vol.30, pp.472-478, 2012.

L. Barbour, C. Mccurdy, T. Hernandez, J. Kirwan, P. Catalano et al., Cellular Mechanisms for Insulin Resistance in Normal Pregnancy and Gestational Diabetes, Diabetes Care, vol.30, issue.Supplement 2, pp.112-119, 2007.
DOI : 10.2337/dc07-s202

P. Catalano, L. Presley, and J. Minium, Fetuses of Obese Mothers Develop Insulin Resistance in Utero, Diabetes Care, vol.32, issue.6, pp.1076-1080, 2009.
DOI : 10.2337/dc08-2077

O. Langer, Fetal Macrosomia: Etiologic Factors, Clinical Obstetrics and Gynecology, vol.43, issue.2, pp.283-297, 2000.
DOI : 10.1097/00003081-200006000-00006

I. Olsen, S. Groveman, M. Lawson, R. Clark, and B. Zemel, New Intrauterine Growth Curves Based on United States Data, PEDIATRICS, vol.125, issue.2, pp.214-224, 2010.
DOI : 10.1542/peds.2009-0913

K. Piper, S. Brickwood, L. Turnpenny, I. Cameron, S. Ball et al., Beta cell differentiation during early human pancreas development, Journal of Endocrinology, vol.181, issue.1, pp.11-23, 2004.
DOI : 10.1677/joe.0.1810011

C. Gicquel and B. Le, Hormonal Regulation of Fetal Growth, Hormone Research in Paediatrics, vol.65, issue.3, pp.28-33, 2006.
DOI : 10.1159/000091503

A. Smerieri, M. Petraroli, M. Ziveri, C. Volta, S. Bernasconi et al., Effects of Cord Serum Insulin, IGF-II, IGFBP-2, IL-6 and Cortisol Concentrations on Human Birth Weight and Length: Pilot Study, PLoS ONE, vol.341, issue.12, p.29562, 2011.
DOI : 10.1371/journal.pone.0029562.t004

Z. Yu, S. Han, J. Zhu, X. Sun, C. Ji et al., Pre-Pregnancy Body Mass Index in Relation to Infant Birth Weight and Offspring Overweight/Obesity: A Systematic Review and Meta-Analysis, PLoS ONE, vol.9, issue.2, p.61627, 2013.
DOI : 10.1371/journal.pone.0061627.s011

J. Dashe, D. Mcintire, M. Lucas, and K. Leveno, Effects of symmetric and asymmetric fetal growth on pregnancy outcomes, Obstet Gynecol, vol.96, pp.321-327, 2000.

H. Lipkind, A. Curry, M. Huynh, L. Thorpe, and T. Matte, Birth Outcomes Among Offspring of Women Exposed to the September 11, 2001, Terrorist Attacks, Obstetrics & Gynecology, vol.116, issue.4, pp.917-925, 2001.
DOI : 10.1097/AOG.0b013e3181f2f6a2

J. Mairesse, J. Lesage, C. Breton, B. Breant, T. Hahn et al., Maternal stress alters endocrine function of the feto-placental unit in rats, AJP: Endocrinology and Metabolism, vol.292, issue.6, pp.1526-1533, 2007.
DOI : 10.1152/ajpendo.00574.2006

I. Buhimschi, G. Saade, K. Chwalisz, and R. Garfield, The nitric oxide pathway in pre-eclampsia: pathophysiological implications, Human Reproduction Update, vol.4, issue.1, pp.25-42, 1998.
DOI : 10.1093/humupd/4.1.25

L. Thaete, D. Kushner, E. Dewey, and M. Neerhof, Endothelin and the regulation of uteroplacental perfusion in nitric oxide synthase inhibition-induced fetal growth restriction, Placenta, vol.26, issue.2-3, pp.242-250, 2005.
DOI : 10.1016/j.placenta.2004.06.003

I. Ghalayini, Nitric oxide???cyclic GMP pathway with some emphasis on cavernosal contractility, International Journal of Impotence Research, vol.16, issue.6, pp.459-469, 2004.
DOI : 10.1038/sj.ijir.3901256

N. Winer, B. Branger, E. Azria, V. Tsatsaris, H. Philippe et al., l-Arginine treatment for severe vascular fetal intrauterine growth restriction: A randomized double-bind controlled trial, Clinical Nutrition, vol.28, issue.3, pp.243-248, 2009.
DOI : 10.1016/j.clnu.2009.03.007

J. Walsh and F. Mcauliffe, Prediction and prevention of the macrosomic fetus, European Journal of Obstetrics & Gynecology and Reproductive Biology, vol.162, issue.2, pp.125-130, 2012.
DOI : 10.1016/j.ejogrb.2012.03.005

A. Ornoy, Prenatal origin of obesity and their complications: Gestational diabetes, maternal overweight and the paradoxical effects of fetal growth restriction and macrosomia, Reproductive Toxicology, vol.32, issue.2, pp.205-212, 2011.
DOI : 10.1016/j.reprotox.2011.05.002

M. Gamborg, L. Byberg, F. Rasmussen, P. Andersen, J. Baker et al., Birth Weight and Systolic Blood Pressure in Adolescence and Adulthood: Meta-Regression Analysis of Sex- and Age-specific Results from 20 Nordic Studies, American Journal of Epidemiology, vol.166, issue.6, pp.634-645, 2007.
DOI : 10.1093/aje/kwm042

V. Monti, J. Carlson, S. Hunt, and T. Adams, Relationship of Ghrelin and Leptin Hormones with Body Mass Index and Waist Circumference in a Random Sample of Adults, Journal of the American Dietetic Association, vol.106, issue.6, pp.822-828, 2006.
DOI : 10.1016/j.jada.2006.03.015

H. Christou, J. Connors, M. Ziotopoulou, V. Hatzidakis, E. Papathanassoglou et al., Cord Blood Leptin and Insulin-Like Growth Factor Levels are Independent Predictors of Fetal Growth, The Journal of Clinical Endocrinology & Metabolism, vol.86, issue.2, pp.935-938, 2001.
DOI : 10.1210/jcem.86.2.7217

A. Kourtis, A. Gkiomisi, M. Mouzaki, K. Makedou, A. Anastasilakis et al., Apelin levels in normal pregnancy, Clinical Endocrinology, vol.20, issue.3, pp.367-371, 2011.
DOI : 10.1111/j.1365-2265.2011.04061.x

S. Pitkin, J. Maguire, T. Bonner, and A. Davenport, International Union of Basic and Clinical Pharmacology. LXXIV. Apelin Receptor Nomenclature, Distribution, Pharmacology, and Function, Pharmacological Reviews, vol.62, issue.3, pp.331-342, 2010.
DOI : 10.1124/pr.110.002949

C. Llorens-cortes and A. Beaudet, L???ap??line, un inhibiteur naturel de l???effet antidiur??tique de la vasopressine, m??decine/sciences, vol.21, issue.8-9, pp.741-746, 2005.
DOI : 10.1051/medsci/2005218-9741

M. Kleinz and A. Davenport, Emerging roles of apelin in biology and medicine, Pharmacology & Therapeutics, vol.107, issue.2, pp.198-211, 2005.
DOI : 10.1016/j.pharmthera.2005.04.001

K. Tatemoto, M. Hosoya, Y. Habata, R. Fujii, T. Kakegawa et al., Isolation and Characterization of a Novel Endogenous Peptide Ligand for the Human APJ Receptor, Biochemical and Biophysical Research Communications, vol.251, issue.2, pp.471-476, 1998.
DOI : 10.1006/bbrc.1998.9489

C. Vickers, P. Hales, V. Kaushik, L. Dick, J. Gavin et al., Hydrolysis of Biological Peptides by Human Angiotensin-converting Enzyme-related Carboxypeptidase, Journal of Biological Chemistry, vol.277, issue.17, pp.14838-14843, 2002.
DOI : 10.1074/jbc.M200581200

E. Messari, S. Iturrioz, X. Fassot, C. De, M. Roesch et al., Functional dissociation of apelin receptor signaling and endocytosis: implications for the effects of apelin on arterial blood pressure, Journal of Neurochemistry, vol.90, issue.6, pp.1290-1301, 2004.
DOI : 10.1111/j.1471-4159.2004.02591.x

URL : https://hal.archives-ouvertes.fr/inserm-00424423

O. Dowd, B. Heiber, M. Chan, A. Heng, H. Tsui et al., A human gene that shows identity with the gene encoding the angiotensin receptor is located on chromosome 11, Gene, vol.136, issue.1-2, pp.355-360, 1993.
DOI : 10.1016/0378-1119(93)90495-O

A. Medhurst, C. Jennings, M. Robbins, R. Davis, C. Ellis et al., Pharmacological and immunohistochemical characterization of the APJ receptor and its endogenous ligand apelin, Journal of Neurochemistry, vol.84, issue.5, pp.1162-1172, 2003.
DOI : 10.1046/j.1471-4159.2003.01587.x

B. Masri, B. Knibiehler, and Y. Audigier, Apelin signalling: a promising pathway from cloning to pharmacology, Cellular Signalling, vol.17, issue.4, pp.415-426, 2005.
DOI : 10.1016/j.cellsig.2004.09.018

URL : https://hal.archives-ouvertes.fr/inserm-00481022

N. Zhou, X. Zhang, X. Fan, E. Argyris, J. Fang et al., The N-terminal domain of APJ, a CNS-based coreceptor for HIV-1, is essential for its receptor function and coreceptor activity, Virology, vol.317, issue.1, pp.84-94, 2003.
DOI : 10.1016/j.virol.2003.08.026

D. Lee, S. Ferguson, S. George, O. Dowd, and B. , The fate of the internalized apelin receptor is determined by different isoforms of apelin mediating differential interaction with ??-arrestin, Biochemical and Biophysical Research Communications, vol.395, issue.2, pp.185-189, 2010.
DOI : 10.1016/j.bbrc.2010.03.151

M. Hosoya, Y. Kawamata, S. Fukusumi, R. Fujii, Y. Habata et al., Molecular and Functional Characteristics of APJ. TISSUE DISTRIBUTION OF mRNA AND INTERACTION WITH THE ENDOGENOUS LIGAND APELIN, Journal of Biological Chemistry, vol.275, issue.28, pp.21061-21067, 2000.
DOI : 10.1074/jbc.M908417199

C. Dray, C. Knauf, D. Daviaud, A. Waget, J. Boucher et al., Apelin Stimulates Glucose Utilization in Normal and Obese Insulin-Resistant Mice, Cell Metabolism, vol.8, issue.5, pp.437-445, 2008.
DOI : 10.1016/j.cmet.2008.10.003

URL : https://hal.archives-ouvertes.fr/inserm-00408948

P. Yue, H. Jin, M. Aillaud, A. Deng, J. Azuma et al., Apelin is necessary for the maintenance of insulin sensitivity, AJP: Endocrinology and Metabolism, vol.298, issue.1, pp.59-67, 2010.
DOI : 10.1152/ajpendo.00385.2009

J. Zhong, X. Yu, Y. Huang, L. Yung, C. Lau et al., Apelin modulates aortic vascular tone via endothelial nitric oxide synthase phosphorylation pathway in diabetic mice, Cardiovascular Research, vol.74, issue.3, pp.388-395, 2007.
DOI : 10.1016/j.cardiores.2007.02.002

J. Ishida, T. Hashimoto, Y. Hashimoto, S. Nishiwaki, T. Iguchi et al., Regulatory Roles for APJ, a Seven-transmembrane Receptor Related to Angiotensin-type 1 Receptor in Blood Pressure in Vivo, Journal of Biological Chemistry, vol.279, issue.25, pp.26274-26279, 2004.
DOI : 10.1074/jbc.M404149200

S. Xu, P. Han, M. Huang, J. Wu, C. Chang et al., In vivo, ex vivo, and in vitro studies on apelin's effect on myocardial glucose uptake, Peptides, vol.37, issue.2, pp.320-326, 2012.
DOI : 10.1016/j.peptides.2012.08.004

C. Dray, C. Foussal, C. Attané, O. Kunduzova, D. Daviaud et al., apeline: de la fonction cardiaque au métabolisme énergétique

D. Hardie, The AMP-activated protein kinase pathway - new players upstream and downstream, Journal of Cell Science, vol.117, issue.23, pp.5479-5487, 2004.
DOI : 10.1242/jcs.01540

J. Boucher, B. Masri, D. Daviaud, S. Gesta, C. Guigne et al., Apelin, a Newly Identified Adipokine Up-Regulated by Insulin and Obesity, Endocrinology, vol.146, issue.4, pp.1764-1771, 2005.
DOI : 10.1210/en.2004-1427

URL : https://hal.archives-ouvertes.fr/inserm-00480981

C. Ringstrom, M. Nitert, H. Bennet, M. Fex, P. Valet et al., Apelin is a novel islet peptide, Regulatory Peptides, vol.162, issue.1-3, pp.44-51, 2010.
DOI : 10.1016/j.regpep.2010.03.005

URL : https://hal.archives-ouvertes.fr/inserm-00506142

I. Castan-laurell, C. Dray, C. Knauf, O. Kunduzova, and P. Valet, Apelin, a promising target for type 2 diabetes treatment?, Trends in Endocrinology & Metabolism, vol.23, issue.5, pp.234-241, 2012.
DOI : 10.1016/j.tem.2012.02.005

URL : https://hal.archives-ouvertes.fr/inserm-00756499

W. Sorhede, C. Magnusson, and B. Ahren, The apj receptor is expressed in pancreatic islets and its ligand, apelin, inhibits insulin secretion in mice, Regulatory Peptides, vol.131, issue.1-3, pp.12-17, 2005.
DOI : 10.1016/j.regpep.2005.05.004

I. Castan-laurell, C. Dray, C. Attane, T. Duparc, C. Knauf et al., Apelin, diabetes, and obesity, Endocrine, vol.96, issue.Suppl 1, pp.1-9, 2011.
DOI : 10.1007/s12020-011-9507-9

URL : https://hal.archives-ouvertes.fr/inserm-00617638

D. Daviaud, J. Boucher, S. Gesta, C. Dray, C. Guigne et al., TNF?? up-regulates apelin expression in human and mouse adipose tissue, The FASEB Journal, vol.20, issue.9, pp.1528-1530, 2006.
DOI : 10.1096/fj.05-5243fje

C. Andersen, O. Hilberg, S. Mellemkjaer, J. Nielsen-kudsk, and U. Simonsen, Apelin and pulmonary hypertension, Pulmonary Circulation, vol.1, issue.3, pp.334-346, 2011.
DOI : 10.4103/2045-8932.87299

I. Castan-laurell, M. Vitkova, D. Daviaud, C. Dray, M. Kovacikova et al., Effect of hypocaloric diet-induced weight loss in obese women on plasma apelin and adipose tissue expression of apelin and APJ., European Journal of Endocrinology, vol.158, issue.6
DOI : 10.1530/EJE-08-0039

URL : https://hal.archives-ouvertes.fr/inserm-00277309

J. Jia, Y. Tian, Z. Cao, L. Tao, X. Zhang et al., The polymorphisms of UCP1 genes associated with fat metabolism, obesity and diabetes, Molecular Biology Reports, vol.80, issue.2, pp.1513-1522, 2010.
DOI : 10.1007/s11033-009-9550-2

K. Higuchi, T. Masaki, K. Gotoh, S. Chiba, I. Katsuragi et al., Apelin, an APJ Receptor Ligand, Regulates Body Adiposity and Favors the Messenger Ribonucleic Acid Expression of Uncoupling Proteins in Mice, Endocrinology, vol.148, issue.6, pp.2690-2697, 2007.
DOI : 10.1210/en.2006-1270

A. Reaux, M. De, I. Skultetyova, Z. Lenkei, M. El et al., Physiological role of a novel neuropeptide, apelin, and its receptor in the rat brain, Journal of Neurochemistry, vol.473, issue.4, pp.1085-1096, 2001.
DOI : 10.1046/j.1471-4159.2001.00320.x

G. Pope, E. Roberts, S. Lolait, O. Carroll, and A. , Central and peripheral apelin receptor distribution in the mouse: Species differences with rat, Peptides, vol.33, issue.1, pp.139-148, 2012.
DOI : 10.1016/j.peptides.2011.12.005

J. Stockand, Vasopressin regulation of renal sodium excretion, Kidney International, vol.78, issue.9, pp.849-856, 2010.
DOI : 10.1038/ki.2010.276

A. Valle, N. Hoggard, A. Adams, P. Roca, and J. Speakman, Chronic central administration of apelin-13 over 10 days increases food intake, body weight, locomotor activity and body temperature in C57BL/6 mice, J Neuroendocrinol, vol.20, pp.79-84, 2008.

D. Sunter, A. Hewson, and S. Dickson, Intracerebroventricular injection of apelin-13 reduces food intake in the rat, Neuroscience Letters, vol.353, issue.1, pp.1-4, 2003.
DOI : 10.1016/S0304-3940(03)00351-3

O. Shea, M. Hansen, M. Tatemoto, K. Morris, and M. , Inhibitory Effect of Apelin-12 on Nocturnal Food Intake in the Rat, Nutritional Neuroscience, vol.6, issue.3, pp.163-167, 2003.
DOI : 10.1016/S0167-0115(01)00236-1

R. Goazigo, L. Bodineau, M. De, L. Jeandel, N. Chartrel et al., Apelin and the proopiomelanocortin system: a new regulatory pathway of hypothalamic ??-MSH release, AJP: Endocrinology and Metabolism, vol.301, issue.5, pp.955-966, 2011.
DOI : 10.1152/ajpendo.00090.2011

E. Devic, L. Paquereau, P. Vernier, B. Knibiehler, and Y. Audigier, Expression of a new G protein-coupled receptor X-msr is associated with an endothelial lineage in Xenopus laevis, Mechanisms of Development, vol.59, issue.2, pp.129-140, 1996.
DOI : 10.1016/0925-4773(96)00585-0

A. Japp and D. Newby, The apelin???APJ system in heart failure, Biochemical Pharmacology, vol.75, issue.10, pp.1882-1892, 2008.
DOI : 10.1016/j.bcp.2007.12.015

A. Japp, N. Cruden, D. Amer, V. Li, E. Goudie et al., Vascular Effects of Apelin In Vivo in Man, Journal of the American College of Cardiology, vol.52, issue.11, pp.908-913, 2008.
DOI : 10.1016/j.jacc.2008.06.013

J. Zhong, Y. Huang, L. Yung, C. Lau, F. Leung et al., The novel peptide apelin regulates intrarenal artery tone in diabetic mice, Regulatory Peptides, vol.144, issue.1-3, pp.109-114, 2007.
DOI : 10.1016/j.regpep.2007.06.010

K. Siddiquee, J. Hampton, D. Mcanally, L. May, and L. Smith, The apelin receptor inhibits the angiotensin II type 1 receptor via allosteric trans-inhibition, British Journal of Pharmacology, vol.65, issue.Suppl. 1, pp.1104-1117, 2013.
DOI : 10.1111/j.1476-5381.2012.02192.x

I. Falcao-pires and A. Leite-moreira, Apelin: a novel neurohumoral modulator of the cardiovascular system. Pathophysiologic importance and potential use as a therapeutic target 5, Rev Port Cardiol, vol.24, pp.1263-1276, 2005.

I. Szokodi, P. Tavi, G. Foldes, S. Voutilainen-myllyla, M. Ilves et al., Apelin, the Novel Endogenous Ligand of the Orphan Receptor APJ, Regulates Cardiac Contractility, Circulation Research, vol.91, issue.5, pp.434-440, 2002.
DOI : 10.1161/01.RES.0000033522.37861.69

J. Zhang, C. Ren, Y. Qi, L. Lou, L. Chen et al., Exercise training promotes expression of apelin and APJ of cardiovascular tissues in spontaneously hypertensive rats, Life Sciences, vol.79, issue.12, pp.1153-1159, 2006.
DOI : 10.1016/j.lfs.2006.03.040

K. Chong, R. Gardner, J. Morton, E. Ashley, and T. Mcdonagh, Plasma concentrations of the novel peptide apelin are decreased in patients with chronic heart failure, European Journal of Heart Failure, vol.94, issue.3, pp.355-360, 2006.
DOI : 10.1016/j.ejheart.2005.10.007

R. Ladeiras-lopes, J. Ferreira-martins, and A. Leite-moreira, The apelinergic system: the role played in human physiology and pathology and potential therapeutic applications 2, Arq Bras Cardiol, vol.90, pp.343-349, 2008.

A. Kasai, N. Shintani, M. Oda, M. Kakuda, H. Hashimoto et al., Apelin is a novel angiogenic factor in retinal endothelial cells, Biochemical and Biophysical Research Communications, vol.325, issue.2, pp.395-400, 2004.
DOI : 10.1016/j.bbrc.2004.10.042

O. Cleaver, K. Tonissen, M. Saha, and P. Krieg, Neovascularization of the Xenopus embryo, Developmental Dynamics, vol.210, issue.1, pp.66-77, 1997.
DOI : 10.1002/(SICI)1097-0177(199709)210:1<66::AID-AJA7>3.3.CO;2-S

A. Malamitsi-puchner, D. Gourgiotis, M. Boutsikou, S. Baka, D. Hassiakos et al., Circulating apelin concentrations in mother/infant pairs at term, Acta Paediatrica, vol.11, issue.12, pp.1751-1754, 2007.
DOI : 10.1046/j.1365-2265.1999.00761.x

E. Devic, K. Rizzoti, S. Bodin, B. Knibiehler, and Y. Audigier, Amino acid sequence and embryonic expression of msr/apj, the mouse homolog of Xenopus X-msr and human APJ, Mechanisms of Development, vol.84, issue.1-2, pp.199-203, 1999.
DOI : 10.1016/S0925-4773(99)00081-7

A. Moynihan, M. Hehir, S. Glavey, T. Smith, and J. Morrison, Inhibitory effect of leptin on human uterine contractility in vitro, American Journal of Obstetrics and Gynecology, vol.195, issue.2, pp.504-509, 2006.
DOI : 10.1016/j.ajog.2006.01.106

M. Hehir, S. Glavey, and J. Morrison, Uterorelaxant effect of ghrelin on human myometrial contractility 10, Am J Obstet Gynecol, vol.198, pp.323-325, 2008.

M. Hehir and J. Morrison, The adipokine apelin and human uterine contractility 19, Am J Obstet Gynecol, vol.206, pp.359-364, 2012.

C. Dray, Y. Sakar, C. Vinel, D. Daviaud, B. Masri et al., The Intestinal Glucose???Apelin Cycle Controls Carbohydrate Absorption in Mice, Gastroenterology, vol.144, issue.4, pp.771-780, 2013.
DOI : 10.1053/j.gastro.2013.01.004

URL : https://hal.archives-ouvertes.fr/inserm-00805173

M. Vickers, Developmental programming of the metabolic syndrome - critical windows for intervention, World Journal of Diabetes, vol.2, issue.9, pp.137-148, 2011.
DOI : 10.4239/wjd.v2.i9.137

D. Barker, O. C. Golding, J. Kuh, D. Wadsworth, and M. , Growth in utero, blood pressure in childhood and adult life, and mortality from cardiovascular disease., BMJ, vol.298, issue.6673, pp.564-567, 1989.
DOI : 10.1136/bmj.298.6673.564

C. Hales and D. Barker, Type 2 (non-insulin-dependent) diabetes mellitus: the thrifty phenotype hypothesis, Diabetologia, vol.133, issue.7, pp.595-601, 1992.
DOI : 10.1007/BF00400248

D. Barker, C. Hales, C. Fall, O. C. Phipps, K. Clark et al., Type 2 (non-insulin-dependent) diabetes mellitus, hypertension and hyperlipidaemia (syndrome X): relation to reduced fetal growth, Diabetologia, vol.340, issue.1, pp.62-67, 1993.
DOI : 10.1007/BF00399095

K. Alberti, P. Zimmet, and J. Shaw, Metabolic syndrome-a new world-wide definition. A Consensus Statement from the International Diabetes Federation, Diabetic Medicine, vol.46, issue.5, pp.469-480, 2006.
DOI : 10.1001/jama.290.4.486

K. Gatford, R. Simmons, D. Blasio, M. Robinson, J. Owens et al., Review: Placental Programming of Postnatal Diabetes and Impaired Insulin Action after IUGR, Placenta, vol.31, pp.60-65, 2010.
DOI : 10.1016/j.placenta.2009.12.015

C. Hales and D. Barker, The thrifty phenotype hypothesis, British Medical Bulletin, vol.60, issue.1, pp.5-20, 2001.
DOI : 10.1093/bmb/60.1.5

D. Barker, Developmental origins of adult health and disease, Journal of Epidemiology & Community Health, vol.58, issue.2, pp.114-115, 2004.
DOI : 10.1136/jech.58.2.114

S. Cianfarani, D. Germani, and F. Branca, Low birthweight and adult insulin resistance: the "catch-up growth" hypothesis, Archives of Disease in Childhood - Fetal and Neonatal Edition, vol.81, issue.1, pp.71-73, 1999.
DOI : 10.1136/fn.81.1.F71

A. Dulloo, Thrifty energy metabolism in catch-up growth trajectories to insulin and leptin resistance, Best Practice & Research Clinical Endocrinology & Metabolism, vol.22, issue.1, pp.155-171, 2008.
DOI : 10.1016/j.beem.2007.08.001

C. Hales and S. Ozanne, The dangerous road of catch-up growth, The Journal of Physiology, vol.547, issue.1, pp.5-10, 2003.
DOI : 10.1113/jphysiol.2002.024406

A. Deierlein, A. Siega-riz, K. Chantala, and A. Herring, The Association Between Maternal Glucose Concentration and Child BMI at Age 3 Years, Diabetes Care, vol.34, issue.2, pp.480-484, 2011.
DOI : 10.2337/dc10-1766

B. Grayson, P. Levasseur, S. Williams, M. Smith, D. Marks et al., Changes in Melanocortin Expression and Inflammatory Pathways in Fetal Offspring of Nonhuman Primates Fed a High-Fat Diet, Endocrinology, vol.151, issue.4, pp.1622-1632, 2010.
DOI : 10.1210/en.2009-1019

E. Sullivan, B. Grayson, D. Takahashi, N. Robertson, A. Maier et al., Chronic Consumption of a High-Fat Diet during Pregnancy Causes Perturbations in the Serotonergic System and Increased Anxiety-Like Behavior in Nonhuman Primate Offspring, Journal of Neuroscience, vol.30, issue.10, pp.3826-3830, 2010.
DOI : 10.1523/JNEUROSCI.5560-09.2010

S. Bouret and R. Simerly, Developmental programming of hypothalamic feeding circuits, Clinical Genetics, vol.402, issue.4, pp.295-301, 2006.
DOI : 10.1111/j.1399-0004.2006.00684.x

P. Gluckman, M. Hanson, C. Cooper, and K. Thornburg, Effect of In Utero and Early-Life Conditions on Adult Health and Disease, New England Journal of Medicine, vol.359, issue.1, pp.61-73, 2008.
DOI : 10.1056/NEJMra0708473

W. Kwong, A. Wild, P. Roberts, A. Willis, and T. Fleming, Maternal undernutrition during the preimplantation period of rat development causes blastocyst abnormalities and programming of postnatal hypertension 31, Development, vol.127, pp.4195-4202, 2000.

I. Weaver, N. Cervoni, F. Champagne, D. 'alessio, A. Sharma et al., Epigenetic programming by maternal behavior, Nature Neuroscience, vol.304, issue.8, pp.847-854, 2004.
DOI : 10.1016/S0031-9384(03)00149-5

T. Harder, R. Bergmann, G. Kallischnigg, and A. Plagemann, Duration of Breastfeeding and Risk of Overweight: A Meta-Analysis, American Journal of Epidemiology, vol.162, issue.5, pp.397-403, 2005.
DOI : 10.1093/aje/kwi222

C. Guerrero-bosagna and M. Skinner, Epigenetic Transgenerational Effects of Endocrine Disruptors on Male Reproduction, Seminars in Reproductive Medicine, vol.27, issue.05, pp.403-408, 2009.
DOI : 10.1055/s-0029-1237428

C. Gallou-kabani, A. Gabory, J. Tost, M. Karimi, S. Mayeur et al., Sex- and Diet-Specific Changes of Imprinted Gene Expression and DNA Methylation in Mouse Placenta under a High-Fat Diet, PLoS ONE, vol.142, issue.12, p.14398, 2010.
DOI : 10.1371/journal.pone.0014398.s001

C. Gicquel, A. El-osta, and B. Le, Epigenetic regulation and fetal programming, Best Practice & Research Clinical Endocrinology & Metabolism, vol.22, issue.1, pp.1-16, 2008.
DOI : 10.1016/j.beem.2007.07.009

Z. Altun, S. Uysal, G. Guner, O. Yilmaz, and C. Posaci, Effects of oral L???arginine supplementation on blood pressure and asymmetric dimethylarginine in stress???induced preeclamptic rats, Cell Biochemistry and Function, vol.33, issue.5, pp.648-653, 2008.
DOI : 10.1002/cbf.1491

G. Helmbrecht, M. Farhat, L. Lochbaum, H. Brown, K. Yadgarova et al., l-arginine reverses the adverse pregnancy changes induced by nitric oxide synthase inhibition in the rat, American Journal of Obstetrics and Gynecology, vol.175, issue.4, pp.800-805, 1996.
DOI : 10.1016/S0002-9378(96)80002-0

C. Yallampalli and R. Garfield, Inhibition of nitric oxide synthesis in rats during pregnancy produces signs similar to those of preeclampsia, American Journal of Obstetrics and Gynecology, vol.169, issue.5, pp.1316-1320, 1993.
DOI : 10.1016/0002-9378(93)90299-X

M. Molnar, T. Suto, T. Toth, and F. Hertelendy, Prolonged blockade of nitric oxide synthesis in gravid rats produces sustained hypertension, proteinuria, thrombocytopenia, and intrauterine growth retardation, American Journal of Obstetrics and Gynecology, vol.170, issue.5, pp.1458-1466, 1994.
DOI : 10.1016/S0002-9378(94)70179-2

C. Buhimschi, R. Gokdeniz, G. Saade, I. Buhimschi, and R. Garfield, The effect of chronic nitric oxide synthase inhibition on blood pressure and heart rate in unrestrained pregnant rats as recorded by radiotelemetry, American Journal of Obstetrics and Gynecology, vol.181, issue.1, pp.159-164, 1999.
DOI : 10.1016/S0002-9378(99)70453-9

C. Fernandez, R. Carbajo, and R. Munoz, Prolonged inhibition of nitric oxide synthesis in pregnant rats: effects on blood pressure, fetal growth and litter size, Archives of Gynecology and Obstetrics, vol.133, issue.3, pp.243-248, 2005.
DOI : 10.1007/s00404-004-0633-y

S. Lubarsky, R. Ahokas, S. Friedman, and B. Sibai, The effect of chronic nitric oxide synthesis inhibition on blood pressure and angiotensin II responsiveness in the pregnant rat, American Journal of Obstetrics and Gynecology, vol.176, issue.5, pp.1069-1076, 1997.
DOI : 10.1016/S0002-9378(97)70404-6

G. Osol, C. Barron, N. Gokina, and M. Mandala, Inhibition of Nitric Oxide Synthases Abrogates Pregnancy-Induced Uterine Vascular Expansive Remodeling, Journal of Vascular Research, vol.46, issue.5, pp.478-486, 2009.
DOI : 10.1159/000200963

D. Edwards, C. Arora, D. Bui, and L. Castro, Long-term nitric oxide blockade in the pregnant rat: Effects on blood pressure and plasma levels of endothelin-1, American Journal of Obstetrics and Gynecology, vol.175, issue.2
DOI : 10.1016/S0002-9378(96)70166-7

G. Olson, G. Saade, I. Buhimschi, K. Chwalisz, and R. Garfield, The effect of an endothelin antagonist on blood pressure in a rat model of preeclampsia, American Journal of Obstetrics and Gynecology, vol.181, issue.3, pp.638-641, 1999.
DOI : 10.1016/S0002-9378(99)70506-5

S. Brain, T. Williams, J. Tippins, and H. Morris, Calcitonin gene-related peptide is a potent vasodilator, Nature, vol.270, issue.5997, pp.54-56, 1985.
DOI : 10.1038/313054a0

A. Witlin, P. Gangula, M. Thompson, and C. Yallampalli, Growth and fertility rates in the offspring of pregnant rats treated with L-?? nitro-L-arginine methyl ester (L-NAME), a nitric oxide inhibitor, American Journal of Obstetrics and Gynecology, vol.186, issue.1, pp.89-93, 2002.
DOI : 10.1067/mob.2002.119178

G. Sutton, A. Centanni, and A. Butler, Protein Malnutrition during Pregnancy in C57BL/6J Mice Results in Offspring with Altered Circadian Physiology before Obesity, Endocrinology, vol.151, issue.4, pp.1570-1580, 2010.
DOI : 10.1210/en.2009-1133

R. Khalil and J. Granger, Vascular mechanisms of increased arterial pressure in preeclampsia: lessons from animal models, American Journal of Physiology - Regulatory, Integrative and Comparative Physiology, vol.283, issue.1, pp.29-45, 2002.
DOI : 10.1152/ajpregu.00762.2001

T. Kanda, S. Wakino, K. Homma, K. Yoshioka, S. Tatematsu et al., Rho-kinase as a molecular target for insulin resistance and hypertension, The FASEB Journal, vol.20, pp.169-171, 2006.
DOI : 10.1096/fj.05-4197fje

Y. Hara, S. Wakino, Y. Tanabe, M. Saito, H. Tokuyama et al., Rho and Rho-Kinase Activity in Adipocytes Contributes to a Vicious Cycle in Obesity That May Involve Mechanical Stretch, Science Signaling, vol.4, issue.157, p.3, 2011.
DOI : 10.1126/scisignal.2001227

X. Huang, J. Fu, and W. Wang, The effects of fasudil on the permeability of the rat blood???brain barrier and blood???spinal cord barrier following experimental autoimmune encephalomyelitis, Journal of Neuroimmunology, vol.239, issue.1-2, pp.61-67, 2011.
DOI : 10.1016/j.jneuroim.2011.08.015

F. Delahaye, C. Breton, P. Risold, M. Enache, I. Dutriez-casteloot et al., Maternal Perinatal Undernutrition Drastically Reduces Postnatal Leptin Surge and Affects the Development of Arcuate Nucleus Proopiomelanocortin Neurons in Neonatal Male Rat Pups, Endocrinology, vol.149, issue.2, pp.470-475, 2008.
DOI : 10.1210/en.2007-1263

URL : https://hal.archives-ouvertes.fr/hal-00482224

B. Blondeau, J. Lesage, P. Czernichow, J. Dupouy, and B. Breant, Glucocorticoids impair fetal beta-cell development in rats 1, Am J Physiol Endocrinol Metab, vol.281, pp.592-599, 2001.

J. Seckl, Prenatal glucocorticoids and long-term programming, European Journal of Endocrinology, vol.151, issue.Suppl_3, pp.49-62, 2004.
DOI : 10.1530/eje.0.151U049

E. Jarvie, S. Hauguel-de-mouzon, S. Nelson, N. Sattar, P. Catalano et al., Lipotoxicity in obese pregnancy and its potential role in adverse pregnancy outcome and obesity in the offspring: Figure 1, Clinical Science, vol.86, issue.3, pp.123-129, 2010.
DOI : 10.1056/NEJMoa0706786

S. Arora and A. , Role of neuropeptides in appetite regulation and obesity ??? A review, Neuropeptides, vol.40, issue.6, pp.375-401, 2006.
DOI : 10.1016/j.npep.2006.07.001

K. Kanasaki and D. Koya, Biology of Obesity: Lessons from Animal Models of Obesity, Journal of Biomedicine and Biotechnology, vol.46, issue.5, 2011.
DOI : 10.2337/diabetes.48.11.2189

R. Surwit, C. Kuhn, C. Cochrane, J. Mccubbin, and M. Feinglos, Diet-Induced Type II Diabetes in C57BL/6J Mice, Diabetes, vol.37, issue.9, pp.1163-1167, 1988.
DOI : 10.2337/diab.37.9.1163

M. Winzell and B. Ahren, The High-Fat Diet-Fed Mouse: A Model for Studying Mechanisms and Treatment of Impaired Glucose Tolerance and Type 2 Diabetes, Diabetes, vol.53, issue.Supplement 3, pp.215-219, 2004.
DOI : 10.2337/diabetes.53.suppl_3.S215

C. Dray, C. Debard, J. Jager, E. Disse, D. Daviaud et al., Apelin and APJ regulation in adipose tissue and skeletal muscle of type 2 diabetic mice and humans, AJP: Endocrinology and Metabolism, vol.298, issue.6, pp.1161-1169, 2010.
DOI : 10.1152/ajpendo.00598.2009

URL : https://hal.archives-ouvertes.fr/inserm-00506117

F. Greenwood, J. Landon, and T. Stamp, The plasma sugar, free fatty acid, cortisol, and growth hormone response to insulin. I. In control subjects., Journal of Clinical Investigation, vol.45, issue.4, pp.429-436, 1966.
DOI : 10.1172/JCI105357

A. Reaux-le-goazigo, R. Vear-perez, P. Zizzari, J. Epelbaum, M. Bluet-pajot et al., Cellular localization of apelin and its receptor in the anterior pituitary: evidence for a direct stimulatory action of apelin on ACTH release, AJP: Endocrinology and Metabolism, vol.292, issue.1, pp.7-15, 2007.
DOI : 10.1152/ajpendo.00521.2005

URL : https://hal.archives-ouvertes.fr/inserm-00422910

Y. Liao and F. Qiao, Expression of Apelin in placentas of patients with hypertensive disorders complicating pregnancy] 1, Zhonghua Fu Chan Ke Za Zhi, vol.42, pp.382-385, 2007.

K. Bortoff, C. Qiu, S. Runyon, M. Williams, and R. Maitra, Decreased Maternal Plasma Apelin Concentrations in Preeclampsia, Hypertension in Pregnancy, vol.50, issue.4, pp.398-404, 2012.
DOI : 10.1016/j.regpep.2005.09.032

T. Van-mieghem, B. Van, H. Van, R. Pijnenborg, J. Deprest et al., Maternal Apelin Physiology during Rat Pregnancy: The Role of the Placenta, Placenta, vol.31, issue.8, pp.725-730, 2010.
DOI : 10.1016/j.placenta.2010.06.001

M. Gronberg, R. Amini, M. Stridsberg, E. Janson, and J. Saras, Neuroendocrine markers are expressed in human mammary glands, Regulatory Peptides, vol.160, issue.1-3, pp.68-74, 2010.
DOI : 10.1016/j.regpep.2009.12.011

B. Telejko, M. Kuzmicki, N. Wawrusiewicz-kurylonek, J. Szamatowicz, A. Nikolajuk et al., Plasma apelin levels and apelin/APJ mRNA expression in patients with gestational diabetes mellitus, Diabetes Research and Clinical Practice, vol.87, issue.2, pp.176-183, 2010.
DOI : 10.1016/j.diabres.2009.10.018

H. Inuzuka, H. Nishizawa, A. Inagaki, M. Suzuki, S. Ota et al., Decreased expression of apelin in placentas from severe pre-eclampsia patients, Hypertension in Pregnancy, vol.108, issue.4, 2013.
DOI : 10.1016/j.pharmthera.2005.04.001

L. Cobellis, F. De, A. Mastrogiacomo, D. Giraldi, D. Dattilo et al., Modulation of apelin and APJ receptor in normal and preeclampsiacomplicated placentas 1, Histol Histopathol, vol.22, pp.1-8, 2007.

. Apelin, the natural ligand of the orphan receptor APJ, is abundantly secreted in the colostrum 1, Biochim Biophys Acta, vol.1452, pp.25-35, 1999.

S. Aydin, The presence of the peptides apelin, ghrelin and nesfatin-1 in the human breast milk, and the lowering of their levels in patients with gestational diabetes mellitus, Peptides, vol.31, issue.12, pp.2236-2240, 2010.
DOI : 10.1016/j.peptides.2010.08.021

S. Clee and A. Attie, The Genetic Landscape of Type 2 Diabetes in Mice, Endocrine Reviews, vol.28, issue.1, pp.48-83, 2007.
DOI : 10.1210/er.2006-0035

S. Andrikopoulos, A. Blair, N. Deluca, B. Fam, and J. Proietto, Evaluating the glucose tolerance test in mice, AJP: Endocrinology and Metabolism, vol.295, issue.6, pp.1323-1332, 2008.
DOI : 10.1152/ajpendo.90617.2008

N. Igosheva, A. Abramov, L. Poston, J. Eckert, T. Fleming et al., Maternal Diet-Induced Obesity Alters Mitochondrial Activity and Redox Status in Mouse Oocytes and Zygotes, PLoS ONE, vol.231, issue.2, p.10074, 2010.
DOI : 10.1371/journal.pone.0010074.t003

E. Jungheim, E. Schoeller, K. Marquard, E. Louden, J. Schaffer et al., Diet-Induced Obesity Model: Abnormal Oocytes and Persistent Growth Abnormalities in the Offspring, Endocrinology, vol.151, issue.8, pp.4039-4046, 2010.
DOI : 10.1210/en.2010-0098

K. Luzzo, Q. Wang, S. Purcell, C. M. Jimenez, P. Grindler et al., High Fat Diet Induced Developmental Defects in the Mouse: Oocyte Meiotic Aneuploidy and Fetal Growth Retardation/Brain Defects, PLoS ONE, vol.54, issue.11, p.49217, 2012.
DOI : 10.1371/journal.pone.0049217.g006

L. Butruille, A. Drougard, C. Knauf, E. Moitrot, P. Valet et al., The apelinergic system: Sexual dimorphism and tissue-specific modulations by obesity and insulin resistance in female mice, Peptides, vol.46, pp.94-101, 2013.
DOI : 10.1016/j.peptides.2013.05.013

URL : https://hal.archives-ouvertes.fr/inserm-00851680

M. Heinonen, A. Purhonen, P. Miettinen, M. Paakkonen, E. Pirinen et al., Apelin, orexin-A and leptin plasma levels in morbid obesity and effect of gastric banding, Regulatory Peptides, vol.130, issue.1-2, pp.7-13, 2005.
DOI : 10.1016/j.regpep.2005.05.003

M. Heinonen, D. Laaksonen, T. Karhu, L. Karhunen, T. Laitinen et al., Effect of diet-induced weight loss on plasma apelin and cytokine levels in individuals with the metabolic syndrome, Nutrition, Metabolism and Cardiovascular Diseases, vol.19, issue.9, pp.626-633, 2009.
DOI : 10.1016/j.numecd.2008.12.008

F. Soriguer, L. Garrido-sanchez, S. Garcia-serrano, J. Garcia-almeida, J. Garcia-arnes et al., Apelin Levels Are Increased in Morbidly Obese Subjects with Type 2 Diabetes Mellitus, Obesity Surgery, vol.8, issue.11, pp.1574-1580, 2009.
DOI : 10.1007/s11695-009-9955-y

M. Ramos, M. Crespo-solans, C. Del, J. Cacho, and E. Herrera, Fat accumulation in the rat during early pregnancy is modulated by enhanced insulin responsiveness, American Journal of Physiology - Endocrinology And Metabolism, vol.285, issue.2, pp.318-328, 2003.
DOI : 10.1152/ajpendo.00456.2002

S. Mayeur, Retard de croissance intra-utérin et vulnérabilité au syndrome métabolique : recherche de marqueurs placentaires dans un modèle de dénutrition maternelle prénatale et chez l'Homme, pp.10-11

Y. Kikuchi, M. Yamada, T. Imakiire, T. Kushiyama, K. Higashi et al., A Rho-kinase inhibitor, fasudil, prevents development of diabetes and nephropathy in insulin-resistant diabetic rats, Journal of Endocrinology, vol.192, issue.3, pp.595-603, 2007.
DOI : 10.1677/JOE-06-0045