D. S. Hunter, S. J. Hazel, K. L. Kind, J. A. Owens, J. B. Pitcher et al., Programming the brain: Common outcomes and gaps in knowledge from animal studies of IUGR, Physiol. Behav, vol.164, pp.233-248, 2016.

J. J. Meier, A. E. Butler, Y. Saisho, T. Monchamp, R. Galasso et al., Beta-cell replication is the primary mechanism subserving the postnatal expansion of beta-cell mass in humans, Diabetes, vol.57, pp.1584-1594, 2008.

S. Bonner-weir, Life and death of the pancreatic beta cells, Trends Endocrinol. Metab, vol.11, pp.375-378, 2000.

S. Bonner-weir, C. Aguayo-mazzucato, and G. C. Weir, Dynamic development of the pancreas from birth to adulthood. Ups, J. Med. Sci, vol.121, pp.155-158, 2016.

F. C. Pan and M. Brissova, Pancreas development in humans, Curr. Opin. Endocrinol. Diabetes Obes, vol.21, pp.77-82, 2014.

D. J. Barker, C. N. Hales, C. H. Fall, C. Osmond, K. Phipps et al., Type 2 (non-insulin-dependent) diabetes mellitus, hypertension and hyperlipidaemia (syndrome X): Relation to reduced fetal growth, Diabetologia, vol.36, pp.62-67, 1993.

M. Zheng, K. E. Lamb, C. Grimes, R. Laws, K. Bolton et al., Rapid weight gain during infancy and subsequent adiposity: A systematic review and meta-analysis of evidence, Obes. Rev, vol.19, pp.321-332, 2018.

I. C. Mcmillen, C. L. Adam, and B. S. Muhlhausler, Early origins of obesity: Programming the appetite regulatory system, J. Physiol, vol.565, p.2708, 2005.

M. C. Jorgensen, J. Ahnfelt-ronne, J. Hald, O. D. Madsen, P. Serup et al., An illustrated review of early pancreas development in the mouse, Endocr. Rev, vol.28, pp.685-705, 2007.

M. H. Abdulreda, A. Caicedo, and P. O. Berggren, A Natural Body Window to Study Human Pancreatic Islet Cell Function and Survival. CellR4 Repair Replace, Regen. Reprogram, vol.1, pp.111-122, 2013.

T. Babic and R. A. Travagli, Neural Control of the Pancreas, p.22, 2016.

M. L. Golson and K. H. Kaestner, Epigenetics in formation, function, and failure of the endocrine pancreas, Mol. Metab, vol.6, pp.1066-1076, 2017.

M. M. Rankin and J. A. Kushner, Adaptive beta-cell proliferation is severely restricted with advanced age, Diabetes, vol.58, pp.1365-1372, 2009.

V. S. Moullé, K. Vivot, C. Tremblay, B. Zarrouki, J. Ghislain et al., Glucose and fatty acids synergistically and reversibly promote beta cell proliferation in rats, vol.60, pp.879-888, 2017.

C. Hellerstrom and I. Swenne, Functional maturation and proliferation of fetal pancreatic beta-cells, Diabetes, vol.40, pp.89-93, 1991.

C. Aguayo-mazzucato, A. Koh, I. El-khattabi, W. C. Li, E. Toschi et al., Mafa expression enhances glucose-responsive insulin secretion in neonatal rat beta cells, Diabetologia, vol.54, pp.583-593, 2011.

J. C. Henquin and M. Nenquin, Immaturity of insulin secretion by pancreatic islets isolated from one human neonate, J. Diabetes Investig, vol.9, pp.270-273, 2018.

J. C. Henquin and M. Nenquin, Dynamics and Regulation of Insulin Secretion in Pancreatic Islets from Normal Young Children, PLoS ONE, vol.11, 2016.

C. Bernard-kargar and A. Ktorza, Endocrine pancreas plasticity under physiological and pathological conditions, Diabetes, vol.50, pp.30-35, 2001.

J. H. Nielsen, Beta cell adaptation in pregnancy: A tribute to Claes Hellerstrom, Ups J. Med. Sci, vol.121, pp.151-154, 2016.

L. Scaglia, F. E. Smith, and S. Bonner-weir, Apoptosis contributes to the involution of beta cell mass in the post partum rat pancreas, Endocrinology, vol.136, pp.5461-5468, 1995.

G. Kloppel, M. Lohr, K. Habich, M. Oberholzer, and P. U. Heitz, Islet pathology and the pathogenesis of type 1 and type 2 diabetes mellitus revisited, Surv. Synth. Pathol. Res, vol.4, pp.110-125, 1985.

A. Assmann, K. Ueki, J. N. Winnay, T. Kadowaki, and R. N. Kulkarni, Glucose effects on beta-cell growth and survival require activation of insulin receptors and insulin receptor substrate 2, Mol. Cell. Biol, vol.29, pp.3219-3228, 2009.

T. Okada, C. W. Liew, J. Hu, C. Hinault, M. D. Michael et al., Insulin receptors in beta-cells are critical for islet compensatory growth response to insulin resistance, Proc. Natl. Acad. Sci, vol.104, pp.8977-8982, 2007.

R. E. Stamateris, R. B. Sharma, Y. Kong, P. Ebrahimpour, D. Panday et al., Glucose Induces Mouse beta-Cell Proliferation via IRS2, MTOR, and Cyclin D2 but Not the Insulin Receptor, Diabetes, vol.65, pp.981-995, 2016.

D. L. Curry, L. L. Bennett, and G. M. Grodsky, Dynamics of insulin secretion by the perfused rat pancreas, Endocrinology, vol.83, pp.572-584, 1968.

V. S. Moullé, J. Ghislain, and V. Poitout, Nutrient regulation of pancreatic beta-cell proliferation, Biochimie, vol.143, pp.10-17, 2017.

M. Prentki, F. M. Matschinsky, and S. R. Madiraju, Metabolic signaling in fuel-induced insulin secretion, Cell Metab, vol.18, pp.162-185, 2013.

B. Ahren and J. J. Holst, The cephalic insulin response to meal ingestion in humans is dependent on both cholinergic and noncholinergic mechanisms and is important for postprandial glycemia, Diabetes, vol.50, pp.1030-1038, 2001.

A. Guemes, P. Herrero, J. Bondia, and P. Georgiou, Modeling the effect of the cephalic phase of insulin secretion on glucose metabolism, Med. Biol. Eng. Comput, vol.57, pp.1173-1186, 2019.

Z. Fu, E. R. Gilbert, and D. Liu, Regulation of insulin synthesis and secretion and pancreatic Beta-cell dysfunction in diabetes, Curr. Diabetes Rev, vol.9, pp.25-53, 2013.

J. O. Hill, H. R. Wyatt, and J. C. Peters, The Importance of Energy Balance, Eur. Endocrinol, vol.9, p.2708, 2013.

S. S. Farabi and T. L. Hernandez, Low-Carbohydrate Diets for Gestational Diabetes, Nutrients, vol.11, 1737.

M. O. Duarte-gardea, D. M. Gonzales-pacheco, D. M. Reader, A. M. Thomas, S. R. Wang et al., Academy of Nutrition and Dietetics Gestational Diabetes Evidence-Based Nutrition Practice Guideline, J. Acad. Nutr. Diet, vol.118, pp.1719-1742, 2018.

P. M. Catalano, H. D. Mcintyre, J. K. Cruickshank, D. R. Mccance, A. R. Dyer et al., The hyperglycemia and adverse pregnancy outcome study: Associations of GDM and obesity with pregnancy outcomes, Diabetes Care, vol.35, pp.780-786, 2012.

A. Plagemann, T. Harder, M. Brunn, A. Harder, K. Roepke et al., Hypothalamic proopiomelanocortin promoter methylation becomes altered by early overfeeding: An epigenetic model of obesity and the metabolic syndrome, J. Physiol, vol.587, pp.4963-4976, 2009.

R. N. Beaumont, N. M. Warrington, A. Cavadino, J. Tyrrell, M. Nodzenski et al., Genome-wide association study of offspring birth weight in 86 577 women identifies five novel loci and highlights maternal genetic effects that are independent of fetal genetics, Hum. Mol. Genet, vol.27, pp.742-756, 2018.

P. Zhang, D. Zhu, Y. Zhang, L. Li, X. Chen et al., Synergetic Effects of Prenatal and Postnatal High Sucrose Intake on Glucose Tolerance and Hepatic Insulin Resistance in Rat Offspring, Mol. Nutr. Food Res, vol.62, 2018.

H. Ozkan, S. Topsakal, and O. Ozmen, Investigation of the diabetic effects of maternal high-glucose diet on rats, Biomed. Pharm, vol.110, pp.609-617, 2019.

K. Ishikawa, S. Tsunekawa, M. Ikeniwa, T. Izumoto, A. Iida et al., Long-term pancreatic beta cell exposure to high levels of glucose but not palmitate induces DNA methylation within the insulin gene promoter and represses transcriptional activity, PLoS ONE, vol.10, 2015.

M. Siemelink, A. Verhoef, J. A. Dormans, P. N. Span, and A. H. Piersma, Dietary fatty acid composition during pregnancy and lactation in the rat programs growth and glucose metabolism in the offspring, Diabetologia, vol.45, pp.1397-1403, 2002.

I. Bringhenti, J. A. Moraes-teixeira, M. R. Cunha, F. Ornellas, C. A. Mandarim-de-lacerda et al., Maternal obesity during the preconception and early life periods alters pancreatic development in early and adult life in male mouse offspring, PLoS ONE, vol.8, 2013.

F. Graus-nunes, E. Dalla-corte-frantz, W. R. Lannes, M. C. Da-silva-menezes, C. A. Mandarim-de-lacerda et al., Pregestational maternal obesity impairs endocrine pancreas in male F1 and F2 progeny, Nutrition, vol.31, pp.380-387, 2015.

G. Ailhaud, F. Massiera, P. Weill, P. Legrand, J. M. Alessandri et al., Temporal changes in dietary fats: Role of n-6 polyunsaturated fatty acids in excessive adipose tissue development and relationship to obesity, Prog. Lipid Res, vol.45, pp.203-236, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00126321

B. Koletzko, I. Cetin, and J. T. Brenna, Perinatal Lipid Intake Working, Group. Dietary fat intakes for pregnant and lactating women, Br. J. Nutr, vol.98, pp.873-877, 2007.

P. A. Perez and N. V. Dipatrizio, Impact of maternal western diet-induced obesity on offspring mortality and peripheral endocannabinoid system in mice, PLoS ONE, vol.13, 2018.

B. M. Gregorio, V. Souza-mello, C. A. Mandarim-de-lacerda, and M. B. Aguila, Maternal high-fat diet is associated with altered pancreatic remodelling in mice offspring, Eur. J. Nutr, vol.52, pp.759-769, 2013.

E. Zambrano, T. Sosa-larios, L. Calzada, C. A. Ibanez, C. A. Mendoza-rodriguez et al., Decreased basal insulin secretion from pancreatic islets of pups in a rat model of maternal obesity, J. Endocrinol, vol.231, pp.49-57, 2016.

S. E. Dyrskog, S. Gregersen, and K. Hermansen, High-fat feeding during gestation and nursing period have differential effects on the insulin secretory capacity in offspring from normal Wistar rats, Rev. Diabet Stud, vol.2, pp.136-145, 2005.

P. D. Taylor, J. Mcconnell, I. Y. Khan, K. Holemans, K. M. Lawrence et al., Impaired glucose homeostasis and mitochondrial abnormalities in offspring of rats fed a fat-rich diet in pregnancy, Am. J. Physiol. Regul. Integr. Comp. Physiol, vol.288, pp.134-139, 2005.

Z. Y. Ong and B. S. Muhlhausler, Maternal "junk-food" feeding of rat dams alters food choices and development of the mesolimbic reward pathway in the offspring, FASEB J, vol.25, pp.2167-2179, 2011.

C. Haumaitre, O. Lenoir, and R. Scharfmann, Histone deacetylase inhibitors modify pancreatic cell fate determination and amplify endocrine progenitors, Mol. Cell. Biol, vol.28, pp.6373-6383, 2008.

O. Lenoir, K. Flosseau, F. X. Ma, B. Blondeau, A. Mai et al., Specific control of pancreatic endocrine beta-and delta-cell mass by class IIa histone deacetylases HDAC4, HDAC5, and HDAC9, Diabetes, vol.60, pp.2861-2871, 2011.

J. Julvez, S. Fernandez-barres, F. Gignac, M. Lopez-vicente, M. Bustamante et al., Maternal seafood consumption during pregnancy and child attention outcomes: A cohort study with gene effect modification by PUFA-related genes, Int. J. Epidemiol, 2019.

G. Wu, F. W. Bazer, G. A. Johnson, C. Herring, H. Seo et al., Functional amino acids in the development of the pig placenta, Mol. Reprod. Dev, vol.84, pp.870-882, 2017.

C. M. Herring, F. W. Bazer, G. A. Johnson, and G. Wu, Impacts of maternal dietary protein intake on fetal survival, growth, and development, Exp. Biol. Med, vol.243, pp.525-533, 2018.

M. Kucia, M. Langhammer, S. Gors, E. Albrecht, H. M. Hammon et al., High-protein diet during gestation and lactation affects mammary gland mRNA abundance, milk composition and pre-weaning litter growth in mice, Animal, vol.5, pp.268-277, 2011.

B. Coupe, E. Delamaire, C. Hoebler, I. Grit, P. Even et al., Hypothalamus integrity and appetite regulation in low birth weight rats reared artificially on a high-protein milk formula, J. Nutr. Biochem, vol.22, pp.956-963, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01000454

E. Delamaire, P. Parnet, B. Coupe, C. Hoebler, S. Blat et al., Long term metabolic impact of high protein neonatal feeding: A preliminary study in male rat pups born with a low birth weight, Clin. Nutr, vol.31, pp.741-748, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01415274

K. M. Switkowski, P. F. Jacques, A. Must, M. F. Hivert, A. Fleisch et al., Higher Maternal Protein Intake during Pregnancy Is Associated with Lower Cord Blood Concentrations of Insulin-like Growth Factor (IGF)-II, IGF Binding Protein 3, and Insulin, but Not IGF-I, in a Cohort of Women with High Protein Intake, J. Nutr, vol.147, pp.1392-1400, 2017.

R. Closa-monasterolo, N. Ferre, V. Luque, M. Zaragoza-jordana, V. Grote et al., Sex differences in the endocrine system in response to protein intake early in life, Am. J. Clin. Nutr, vol.94, 1920.

E. Oken, E. Morton-eggleston, S. L. Rifas-shiman, K. M. Switkowski, M. F. Hivert et al., Sex-Specific Associations of Maternal Gestational Glycemia with Hormones in Umbilical Cord Blood at Delivery, Am. J. Perinatol, vol.33, pp.1273-1281, 2016.

D. Sharma, S. Shastri, and P. Sharma, Intrauterine Growth Restriction: Antenatal and Postnatal Aspects, Clin. Med. Insights Pediatr, vol.10, pp.67-83, 2016.

K. M. Godfrey and D. J. Barker, Fetal nutrition and adult disease, Am. J. Clin. Nutr, vol.71, pp.1344-1352, 2000.

C. N. Hales, D. J. Barker, P. M. Clark, L. J. Cox, C. Fall et al., Fetal and infant growth and impaired glucose tolerance at age 64, BMJ, vol.303, pp.1019-1022, 1991.

F. A. Van-assche, L. Aerts, and F. A. De-prins, The fetal endocrine pancreas, Eur. J. Obstet. Gynecol. Reprod. Biol, vol.18, pp.267-272, 1984.

A. Snoeck, C. Remacle, B. Reusens, and J. J. Hoet, Effect of a low protein diet during pregnancy on the fetal rat endocrine pancreas, Biol. Neonate, vol.57, pp.107-118, 1990.

L. Calzada, A. Morales, T. C. Sosa-larios, L. A. Reyes-castro, G. L. Rodriguez-gonzalez et al., Maternal protein restriction during gestation impairs female offspring pancreas development in the rat, Nutr. Res, vol.36, pp.855-862, 2016.

J. H. Park, D. A. Stoffers, R. D. Nicholls, and R. A. Simmons, Development of type 2 diabetes following intrauterine growth retardation in rats is associated with progressive epigenetic silencing of Pdx1, J. Clin. Investig, vol.118, pp.2316-2324, 2008.

T. L. Hernandez, A. Mande, and L. A. Barbour, Nutrition therapy within and beyond gestational diabetes, Diabetes Res. Clin. Pract, vol.145, pp.39-50, 2018.

L. A. Barbour, S. S. Farabi, J. E. Friedman, N. M. Hirsch, M. S. Reece et al., Postprandial Triglycerides Predict Newborn Fat More Strongly than Glucose in Women with Obesity in Early Pregnancy, Obesity, vol.26, pp.1347-1356, 2018.

J. Wang, D. Moore, A. Subramanian, K. K. Cheng, K. A. Toulis et al., Gestational dyslipidaemia and adverse birthweight outcomes: A systematic review and meta-analysis, Obes. Rev, vol.19, pp.1256-1268, 2018.

B. Sears and M. Perry, The role of fatty acids in insulin resistance, Lipids Health Dis, vol.14, 2015.

L. A. Barbour, T. L. Hernandez, F. Lipids, and . Overgrowth, Making Fat from Fat, Clin. Ther, vol.40, pp.1638-1647, 2018.

M. L. Blumfield, A. J. Hure, L. K. Macdonald-wicks, R. Smith, S. J. Simpson et al., Dietary balance during pregnancy is associated with fetal adiposity and fat distribution, Am. J. Clin. Nutr, vol.96, pp.1032-1041, 2012.

G. Wu, B. Imhoff-kunsch, and A. W. Girard, Biological mechanisms for nutritional regulation of maternal health and fetal development, Paediatr. Perinat. Epidemiol, vol.26, pp.4-26, 2012.

B. Coupe, V. Amarger, I. Grit, A. Benani, and P. Parnet, Nutritional programming affects hypothalamic organization and early response to leptin, Endocrinology, vol.151, pp.702-713, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00452851

B. Coupe, I. Grit, D. Darmaun, and P. Parnet, The timing of "catch-up growth" affects metabolism and appetite regulation in male rats born with intrauterine growth restriction, Am. J. Physiol. Regul. Integr. Comp. Physiol, vol.297, pp.813-824, 2009.

A. Martin-agnoux, M. C. Alexandre-gouabau, G. Le-drean, J. P. Antignac, and P. Parnet, Relative contribution of foetal and post-natal nutritional periods on feeding regulation in adult rats, Acta Physiol. (Oxf.), vol.210, pp.188-201, 2014.

A. M. Agnoux, J. P. Antignac, G. Simard, G. Poupeau, D. Darmaun et al., Time window-dependent effect of perinatal maternal protein restriction on insulin sensitivity and energy substrate oxidation in adult male offspring, Am. J. Physiol. Regul. Integr. Comp. Physiol, vol.307, pp.184-197, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01389210

E. D. Frantz, M. B. Aguila, R. Pinheiro-mulder-ada, and C. A. Mandarim-de-lacerda, Transgenerational endocrine pancreatic adaptation in mice from maternal protein restriction in utero, Mech. Ageing Dev, vol.132, pp.110-116, 2011.

E. Bertin, M. N. Gangnerau, D. Bailbe, and B. Portha, Glucose metabolism and beta-cell mass in adult offspring of rats protein and/or energy restricted during the last week of pregnancy, Am. J. Physiol, vol.277, pp.11-17, 1999.

S. Morimoto, L. Calzada, T. C. Sosa, L. A. Reyes-castro, G. L. Rodriguez-gonzalez et al., Emergence of ageing-related changes in insulin secretion by pancreatic islets of male rat offspring of mothers fed a low-protein diet, Br. J. Nutr, vol.107, pp.1562-1565, 2012.

M. P. Barnett, A. R. Phillips, P. M. Harris, and G. J. Cooper, Impaired insulin secretion in perfused pancreases isolated from offspring of female rats fed a low protein whey-based diet, JOP, vol.9, pp.477-488, 2008.

C. A. Monteiro, G. Cannon, R. B. Levy, J. C. Moubarac, M. L. Louzada et al., Ultra-processed foods: What they are and how to identify them, Public Health Nutr, vol.22, pp.936-941, 2019.

J. M. Poti, B. Braga, and B. Qin, Ultra-processed Food Intake and Obesity: What Really Matters for Health-Processing or Nutrient Content?, Curr. Obes. Rep, vol.6, pp.420-431, 2017.

F. Juul, E. Martinez-steele, N. Parekh, C. A. Monteiro, and V. W. Chang, Ultra-processed food consumption and excess weight among US adults, Br. J. Nutr, vol.120, pp.90-100, 2018.

B. Srour, L. K. Fezeu, E. Kesse-guyot, B. Alles, C. Mejean et al., Ultra-processed food intake and risk of cardiovascular disease: Prospective cohort study (NutriNet-Sante), BMJ, vol.365, 1451.
URL : https://hal.archives-ouvertes.fr/hal-02177544

K. W. Rohatgi, R. A. Tinius, W. T. Cade, E. M. Steele, A. G. Cahill et al., Relationships between consumption of ultra-processed foods, gestational weight gain and neonatal outcomes in a sample of US pregnant women, vol.5, 2017.

D. S. Sartorelli, L. C. Crivellenti, D. C. Zuccolotto, and L. J. Franco, Relationship between minimally and ultra-processed food intake during pregnancy with obesity and gestational diabetes mellitus, Cad. Saude Publica, vol.35, 2019.

C. B. Gomes, M. B. Malta, S. J. Papini, M. H. Benicio, J. E. Corrente et al., Adherence to dietary patterns during pregnancy and association with maternal characteristics in pregnant Brazilian women, Nutrition, vol.62, pp.85-92, 2019.

C. B. Gomes, M. B. Malta, M. Louzada, M. H. Benicio, A. J. Barros et al., Ultra-processed Food Consumption by Pregnant Women: The Effect of an Educational Intervention with Health Professionals. Matern, Child Health J, vol.23, pp.692-703, 2019.

F. Staud, R. Karahoda, and . Trophoblast, The central unit of fetal growth, protection and programming, Int. J. Biochem. Cell. Biol, vol.105, pp.35-40, 2018.

K. N. Fontes, M. W. Reginatto, N. L. Silva, C. B. Andrade, F. F. Bloise et al., Dysregulation of placental ABC transporters in a murine model of malaria-induced preterm labor, Sci. Rep, vol.9, 2019.

L. Bordoni, C. Nasuti, A. Di-stefano, L. Marinelli, and R. Gabbianelli, Epigenetic Memory of Early-Life Parental Perturbation: Dopamine Decrease and DNA Methylation Changes in Offspring, Oxid. Med. Cell. Longev, 2019.

C. Nasuti, P. Fattoretti, M. Carloni, D. Fedeli, M. Ubaldi et al., Neonatal exposure to permethrin pesticide causes lifelong fear and spatial learning deficits and alters hippocampal morphology of synapses, J. Neurodev. Disord, vol.6, issue.7, 2014.

M. Vrijheid, M. Casas, M. Gascon, D. Valvi, and M. Nieuwenhuijsen, Environmental pollutants and child health-A review of recent concerns, Int J. Hyg. Environ. Health, vol.219, pp.331-342, 2016.

M. Garcia-arevalo, P. Alonso-magdalena, J. M. Servitja, T. Boronat-belda, B. Merino et al., Maternal Exposure to Bisphenol-A During Pregnancy Increases Pancreatic beta-Cell Growth During Early Life in Male Mice Offspring, Endocrinology, vol.157, pp.4158-4171, 2016.

J. C. Roze, D. Darmaun, C. Y. Boquien, C. Flamant, J. C. Picaud et al., The apparent breastfeeding paradox in very preterm infants: Relationship between breast feeding, early weight gain and neurodevelopment based on results from two cohorts, EPIPAGE and LIFT, BMJ Open, vol.2, 2012.

O. Ballard and A. L. Morrow, Human milk composition: Nutrients and bioactive factors, Pediatr. Clin. N. Am, vol.60, pp.49-74, 2013.

B. Koletzko, C. Agostoni, R. Bergmann, K. Ritzenthaler, and R. Shamir, Physiological aspects of human milk lipids and implications for infant feeding: A workshop report, Acta Paediatr, vol.100, pp.1405-1415, 2011.

N. J. Andreas, B. Kampmann, and K. Mehring-le-doare, Human breast milk: A review on its composition and bioactivity, Early Hum. Dev, vol.91, pp.629-635, 2015.

J. Makela, K. Linderborg, H. Niinikoski, B. Yang, and H. Lagstrom, Breast milk fatty acid composition differs between overweight and normal weight women: The STEPS Study, Eur. J. Nutr, vol.52, pp.727-735, 2013.

A. Martin-agnoux, J. P. Antignac, C. Y. Boquien, A. David, E. Desnots et al., Perinatal protein restriction affects milk free amino acid and fatty acid profile in lactating rats: Potential role on pup growth and metabolic status, J. Nutr. Biochem, vol.26, pp.784-795, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02634633

S. M. Innis, Human milk and formula fatty acids, J. Pediatr, vol.120, pp.56-61, 1992.

P. S. Ranade and S. S. Rao, Maternal long-chain PUFA supplementation during protein deficiency improves brain fatty acid accretion in rat pups by altering the milk fatty acid composition of the dam, J. Nutr. Sci, 2013.