L. Liu, K. Hill, S. Oza, D. Hogan, Y. Chu et al., Levels and Causes of Mortality under Age Five Years, Disease control priorities, pp.71-84, 2015.
DOI : 10.1596/978-1-4648-0348-2_ch4

J. Lawn, S. Cousens, and J. Zupan, 4 million neonatal deaths: When? Where? Why?, The Lancet, vol.365, issue.9462, pp.891-900, 2005.
DOI : 10.1016/S0140-6736(05)71048-5

A. Wilcock, P. Begley, A. Stevens, A. Whatmore, and S. Victor, The metabolomics of necrotising enterocolitis in preterm babies: an exploratory study, The Journal of Maternal-Fetal & Neonatal Medicine, vol.29, issue.5, pp.758-62, 2015.
DOI : 10.1093/bioinformatics/btt703

M. Decaro and N. Vain, Hyperglycaemia in preterm neonates: What to know, what to do, Early Human Development, vol.87, pp.19-22, 2011.
DOI : 10.1016/j.earlhumdev.2011.01.005

K. Sadeghi, A. Berger, M. Langgartner, A. Prusa, M. Hayde et al., Immaturity of Infection Control in Preterm and Term Newborns Is Associated with Impaired Toll???Like Receptor Signaling, The Journal of Infectious Diseases, vol.195, issue.2, pp.296-302, 2007.
DOI : 10.1086/509892

L. Wisgrill, A. Groschopf, E. Herndl, K. Sadeghi, A. Spittler et al., Reduced TNF-?? response in preterm neonates is associated with impaired nonclassic monocyte function, Journal of Leukocyte Biology, vol.100, issue.3, pp.607-619, 2016.
DOI : 10.1189/jlb.4A0116-001RR

E. Forster-waldl, K. Sadeghi, D. Tamandl, B. Gerhold, U. Hallwirth et al., Monocyte Toll-Like Receptor 4 Expression and LPS-Induced Cytokine Production Increase during Gestational Aging, Pediatric Research, vol.21, issue.1, pp.121-125, 2005.
DOI : 10.1016/S1043-4666(02)00498-2

P. Sangild, T. Thymann, M. Schmidt, B. Stoll, D. Burrin et al., Invited Review: The preterm pig as a model in pediatric gastroenterology, Journal of Animal Science, vol.91, issue.10, pp.4713-4742, 2013.
DOI : 10.2527/jas.2013-6359

N. Nanthakumar, C. Young, J. Ko, D. Meng, J. Chen et al., Glucocorticoid responsiveness in developing human intestine: possible role in prevention of necrotizing enterocolitis, AJP: Gastrointestinal and Liver Physiology, vol.288, issue.1, pp.85-92, 2004.
DOI : 10.1152/ajpgi.00169.2004

R. Torrazza, N. Li, and J. Neu, Decoding the enigma of necrotizing enterocolitis in premature infants, Pathophysiology, vol.21, issue.1, pp.21-28, 2014.
DOI : 10.1016/j.pathophys.2013.11.011

J. Neu, Necrotizing Enterocolitis: The Mystery Goes On, Neonatology, vol.106, issue.4, pp.289-95, 2014.
DOI : 10.1159/000365130

T. Garite, R. Clark, and J. Thorp, Intrauterine growth restriction increases morbidity and mortality among premature neonates, American Journal of Obstetrics and Gynecology, vol.191, issue.2, pp.481-488, 2004.
DOI : 10.1016/j.ajog.2004.01.036

J. Morgan, L. Young, and W. Mcguire, Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants, Cochrane Database Syst Rev, vol.doi, pp.10-1002, 2013.

X. Wang, G. Lin, C. Liu, C. Feng, H. Zhou et al., Temporal proteomic analysis reveals defects in small-intestinal development of porcine fetuses with intrauterine growth restriction, The Journal of Nutritional Biochemistry, vol.25, issue.7, pp.785-95, 2014.
DOI : 10.1016/j.jnutbio.2014.03.008

W. Dietz, Nutritional Adaptation of the Gastrointestinal Tract of the Newborn, Nestle Nutrition Workshop Series, 1984.

R. Montgomery, A. Mulberg, and R. Grand, Development of the human gastrointestinal tract: Twenty years of progress, Gastroenterology, vol.116, issue.3, pp.702-733, 1999.
DOI : 10.1016/S0016-5085(99)70193-9

A. Maheshwari, Immunologic and Hematological Abnormalities in Necrotizing Enterocolitis, Clinics in Perinatology, vol.42, issue.3, pp.567-85, 2015.
DOI : 10.1016/j.clp.2015.04.014

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4530495/pdf

C. Young, S. Kingma, and J. Neu, Ischemia-Reperfusion and Neonatal Intestinal Injury, The Journal of Pediatrics, vol.158, issue.2, pp.25-33, 2011.
DOI : 10.1016/j.jpeds.2010.11.009

P. Sangild, M. Schmidt, J. Elnif, C. Bjornvad, B. Westrom et al., Prenatal Development of Gastrointestinal Function in the Pig and the Effects of Fetal Esophageal Obstruction, Pediatric Research, vol.95, issue.3, pp.416-440, 2002.
DOI : 10.1073/pnas.95.5.2117

P. Sangild, A. Fowden, and J. Trahair, How does the foetal gastrointestinal tract develop in preparation for enteral nutrition after birth?, Livestock Production Science, vol.66, issue.2, pp.141-50, 2000.
DOI : 10.1016/S0301-6226(00)00221-9

B. Oosterloo, M. Premkumar, B. Stoll, O. Olutoye, T. Thymann et al., Dual purpose use of preterm piglets as a model of pediatric GI disease, Veterinary Immunology and Immunopathology, vol.159, issue.3-4, pp.3-4156, 2014.
DOI : 10.1016/j.vetimm.2014.02.012

E. Roura, S. Koopmans, J. Lallès, L. Huerou-luron, I. De-jager et al., Abstract, Nutrition Research Reviews, vol.45, issue.01, pp.60-90, 2016.
DOI : 10.1002/erv.2256

L. Canario, E. Cantoni, L. Bihan, E. Caritez, J. Billon et al., Between-breed variability of stillbirth and its relationship with sow and piglet characteristics, Journal of Animal Science, vol.84, issue.12, pp.3185-96, 2006.
DOI : 10.2527/jas.2005-775

L. Canario, M. Père, T. Tribout, F. Thomas, C. David et al., Estimation of genetic trends from 1977 to 1998 of body composition and physiological state of Large White pigs at birth, animal, vol.28, issue.10, pp.1409-1422, 2007.
DOI : 10.1159/000240806

V. Voillet, M. Sancristobal, Y. Lippi, P. Martin, N. Iannuccelli et al., Muscle transcriptomic investigation of late fetal development identifies candidate genes for piglet maturity, BMC Genomics, vol.15, issue.1, p.797, 2014.
DOI : 10.1186/1471-2164-15-797

R. Li, Q. Sun, Y. Jia, R. Cong, Y. Ni et al., Coordinated miRNA/mRNA Expression Profiles for Understanding Breed-Specific Metabolic Characters of Liver between Erhualian and Large White Pigs, PLoS ONE, vol.25, issue.6, p.38716, 2012.
DOI : 10.1371/journal.pone.0038716.s004

M. Père, Materno-foetal exchanges and utilisation of nutrients by the foetus: comparison between species, Reproduction Nutrition Development, vol.43, issue.1, pp.1-15, 2003.
DOI : 10.1051/rnd:2003002

J. Marion, Y. Petersen, V. Romé, F. Thomas, P. Sangild et al., Early Weaning Stimulates Intestinal Brush Border Enzyme Activities in Piglets, Mainly at the Posttranscriptional Level, Journal of Pediatric Gastroenterology and Nutrition, vol.41, issue.4, pp.401-411, 2005.
DOI : 10.1097/01.mpg.0000177704.99786.07

URL : http://pdfs.journals.lww.com/jpgn/2005/10000/Early_Weaning_Stimulates_Intestinal_Brush_Border.5.pdf?token=method|ExpireAbsolute;source|Journals;ttl|1503100645511;payload|mY8D3u1TCCsNvP5E421JYK6N6XICDamxByyYpaNzk7FKjTaa1Yz22MivkHZqjGP4kdS2v0J76WGAnHACH69s21Csk0OpQi3YbjEMdSoz2UhVybFqQxA7lKwSUlA502zQZr96TQRwhVlocEp/sJ586aVbcBFlltKNKo+tbuMfL73hiPqJliudqs17cHeLcLbV/CqjlP3IO0jGHlHQtJWcICDdAyGJMnpi6RlbEJaRheGeh5z5uvqz3FLHgPKVXJzdHuxZtXbLmX77MTrMMtOQeiT8pGNu1wHOW++xqZbOALo=;hash|phCiMT1Xf+AMRn0RdkFaOQ==

O. Lowry, N. Rosebrough, A. Farr, and R. Randall, Protein measurement with the Folin phenol reagent, J Biol Chem, vol.193, issue.1, pp.265-75, 1951.

D. Inca, R. Kloareg, M. Gras-le-guen, C. , L. Huërou-luron et al., Intrauterine Growth Restriction Modifies the Developmental Pattern of Intestinal Structure, Transcriptomic Profile, and Bacterial Colonization in Neonatal Pigs, Journal of Nutrition, vol.140, issue.5, pp.925-956, 2010.
DOI : 10.3945/jn.109.116822

J. Vandesompele, D. Preter, K. Pattyn, F. Poppe, B. Van-roy et al., Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes, Genome Biol, vol.3, issue.7, p.34, 2002.

R. Development and C. Team, R: A Language and Environment for Statistical Computing. Vienna, Austria: the R Foundation for Statistical Computing, 2013.

D. Tabas-madrid, R. Nogales-cadenas, and A. Pascual-montano, GeneCodis3: a non-redundant and modular enrichment analysis tool for functional genomics, Nucleic Acids Research, vol.40, issue.W1, pp.478-83, 2012.
DOI : 10.1093/nar/gks402

URL : https://academic.oup.com/nar/article-pdf/40/W1/W478/4913775/gks402.pdf

A. Krämer, J. Green, J. Pollard, . Jr, and S. Tugendreich, Causal analysis approaches in Ingenuity Pathway Analysis, Bioinformatics, vol.30, issue.4, pp.523-553, 2014.
DOI : 10.1093/bioinformatics/btt703

L. Cao, K. González, I. Déjean, and S. , integrOmics: an R package to unravel relationships between two omics datasets, Bioinformatics, vol.25, issue.21, pp.2855-2861, 2009.
DOI : 10.1093/bioinformatics/btp515

L. Cao, K. Rossouw, D. Robert-granie, C. Besse, and P. , A sparse PLS for variable selection when integrating omics data, Statistical Applications in Genetics and Molecular Biology, vol.7, issue.1
URL : https://hal.archives-ouvertes.fr/hal-00300204

M. Dash and H. Liu, Feature selection for classification, Intelligent Data Analysis, vol.1, issue.1-4, pp.131-56, 1997.
DOI : 10.1016/S1088-467X(97)00008-5

S. Geisser, The Predictive Sample Reuse Method with Applications, Journal of the American Statistical Association, vol.36, issue.2, pp.320-328, 1975.
DOI : 10.1007/BF02297848

M. Stone, Cross-validatory choice and assessment of statistical predictions, J R Stat Soc Ser B, vol.36, issue.2, pp.111-158, 1974.

G. Randall, L. Ecuyer, and C. , Tissue Glycogen and Blood Glucose and Fructose Levels in the Pig Fetus during the Second Half of Gestation, Neonatology, vol.28, issue.1-2, pp.74-82, 1976.
DOI : 10.1159/000240806

J. Vallet, A. Mn, J. Miles, and B. Freking, Placental accommodations for transport and metabolism during intra-uterine crowding in pigs, Journal of Animal Science and Biotechnology, vol.5, issue.1, pp.55-65, 2014.
DOI : 10.1677/joe.0.1060355

URL : http://doi.org/10.1186/2049-1891-5-55

S. Bischoff, S. Tsai, N. Hardison, A. Motsinger-reif, B. Freking et al., Characterization of Conserved and Nonconserved Imprinted Genes in Swine1, Biology of Reproduction, vol.81, issue.5, pp.906-926, 2009.
DOI : 10.1095/biolreprod.109.078139

K. Byrne, T. Vuocolo, C. Gondro, J. White, N. Cockett et al., A gene network switch enhances the oxidative capacity of ovine skeletal muscle during late fetal development, BMC Genomics, vol.11, issue.1, p.378, 2010.
DOI : 10.1186/1471-2164-11-378

E. Levy and D. Ménard, Developmental aspects of lipid and lipoprotein synthesis and secretion in human gut, Microscopy Research and Technique, vol.6, issue.4, pp.363-73, 2000.
DOI : 10.1016/0005-2760(94)90249-6

J. Leenhouwers, E. Knol, P. De-groot, H. Vos, and T. Van-der-lende, Fetal development in the pig in relation to genetic merit for piglet survival, Journal of Animal Science, vol.80, issue.7, pp.1759-70, 2002.
DOI : 10.2527/2002.8071759x

T. Van-der-lende, E. Knol, and J. Leenhouwers, Prenatal development as a predisposing factor for perinatal losses in pigs, Reprod Suppl, vol.58, pp.247-61, 2001.

P. Jiang, J. Wan, M. Cilieborg, W. Sit, and P. Sangild, Premature Delivery Reduces Intestinal Cytoskeleton, Metabolism, and Stress Response Proteins in Newborn Formula-Fed Pigs, Journal of Pediatric Gastroenterology and Nutrition, vol.56, issue.6, pp.615-637, 2013.
DOI : 10.1097/MPG.0b013e318288cf71

URL : http://pdfs.journals.lww.com/jpgn/2013/06000/Premature_Delivery_Reduces_Intestinal.8.pdf?token=method|ExpireAbsolute;source|Journals;ttl|1503500938400;payload|mY8D3u1TCCsNvP5E421JYK6N6XICDamxByyYpaNzk7FKjTaa1Yz22MivkHZqjGP4kdS2v0J76WGAnHACH69s21Csk0OpQi3YbjEMdSoz2UhVybFqQxA7lKwSUlA502zQZr96TQRwhVlocEp/sJ586aVbcBFlltKNKo+tbuMfL73hiPqJliudqs17cHeLcLbV/CqjlP3IO0jGHlHQtJWcICDdAyGJMnpi6RlbEJaRheGeh5z5uvqz3FLHgPKVXJzdCvqZ1PRW9YMwo89+DBVWTpSfNBe8YJOPqdwMEqLivRc=;hash|dSs+Xm7/HcLLJzlbO4cq/g==

D. Espinoza, L. Boros, S. Crunkhorn, H. Gami, and M. Patti, Dual modulation of both lipid oxidation and synthesis by peroxisome proliferator-activated receptor-?? coactivator-1?? and -1?? in cultured myotubes, The FASEB Journal, vol.24, issue.4, pp.1003-1017, 2010.
DOI : 10.1096/fj.09-133728

M. Chan and Z. Arany, The many roles of PGC-1?? in muscle ??? recent developments, Metabolism, vol.63, issue.4, pp.441-51, 2014.
DOI : 10.1016/j.metabol.2014.01.006

S. Koo, H. Satoh, S. Herzig, C. Lee, S. Hedrick et al., PGC-1 promotes insulin resistance in liver through PPAR-??-dependent induction of TRB-3, Nature Medicine, vol.50, issue.5, pp.530-534, 2004.
DOI : 10.2337/diabetes.50.10.2316

R. Zoncu, A. Efeyan, and D. Sabatini, mTOR: from growth signal integration to cancer, diabetes and ageing, Nature Reviews Molecular Cell Biology, vol.20, issue.1, pp.21-35, 2011.
DOI : 10.4161/auto.5618

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3390257/pdf

E. Voudoukis, K. Karmiris, and I. Koutroubakis, Multipotent role of platelets in inflammatory bowel diseases: A clinical approach, World Journal of Gastroenterology, vol.20, issue.12, pp.3180-90, 2014.
DOI : 10.3748/wjg.v20.i12.3180

S. Zhao, D. Kuhar, J. Lunney, H. Dawson, C. Guidry et al., Gene expression profiling in Salmonella Choleraesuis-infected porcine lung using a long oligonucleotide microarray, Mammalian Genome, vol.86, issue.Pt 1, pp.777-89, 2006.
DOI : 10.2527/2004.8251261x

H. Van-gorp, P. Delputte, and H. Nauwynck, Scavenger receptor CD163, a Jack-of-all-trades and potential target for cell-directed therapy, Molecular Immunology, vol.47, issue.7-8, pp.7-81650, 2010.
DOI : 10.1016/j.molimm.2010.02.008

A. Maheshwari, D. Kelly, T. Nicola, N. Ambalavanan, S. Jain et al., TGF-??2 Suppresses Macrophage Cytokine Production and Mucosal Inflammatory Responses in the Developing Intestine, Gastroenterology, vol.140, issue.1, pp.242-53, 2011.
DOI : 10.1053/j.gastro.2010.09.043

L. Smythies, M. Sellers, R. Clements, M. Mosteller-barnum, G. Meng et al., Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity, Journal of Clinical Investigation, vol.115, issue.1, pp.66-75, 2005.
DOI : 10.1172/JCI200519229

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC539188/pdf

S. Rautava, L. Lu, N. Nanthakumar, A. Dubert-ferrandon, and W. Walker, TGF-??2 Induces Maturation of Immature Human Intestinal Epithelial Cells and Inhibits Inflammatory Cytokine Responses Induced Via the NF-??B Pathway, Journal of Pediatric Gastroenterology and Nutrition, vol.54, issue.5, pp.630-638, 2012.
DOI : 10.1097/MPG.0b013e31823e7c29

E. Claud, X. Zhang, E. Petrof, and J. Sun, Developmentally regulated tumor necrosis factor-?? induced nuclear factor-??B activation in intestinal epithelium, AJP: Gastrointestinal and Liver Physiology, vol.292, issue.5, pp.1411-1420, 2007.
DOI : 10.1152/ajpgi.00557.2006

URL : http://ajpgi.physiology.org/content/ajpgi/292/5/G1411.full.pdf

M. Lotz, D. Gütle, S. Walther, S. Ménard, C. Bogdan et al., Postnatal acquisition of endotoxin tolerance in intestinal epithelial cells, The Journal of Experimental Medicine, vol.270, issue.4, pp.973-84, 2006.
DOI : 10.1073/pnas.0401710101

P. Sangild, J. Mei, A. Fowden, and R. Xu, The prenatal porcine intestine has low transforming growth factor-beta ligand and receptor density and shows reduced trophic response to enteral diets, AJP: Regulatory, Integrative and Comparative Physiology, vol.296, issue.4, pp.1053-62, 2009.
DOI : 10.1152/ajpregu.90790.2008

URL : http://ajpregu.physiology.org/content/ajpregu/296/4/R1053.full.pdf

R. Derijk, M. Schaaf, and E. De-kloet, Glucocorticoid receptor variants: clinical implications, The Journal of Steroid Biochemistry and Molecular Biology, vol.81, issue.2, pp.103-125, 2002.
DOI : 10.1016/S0960-0760(02)00062-6

H. Zou, R. Li, Y. Jia, X. Yang, Y. Ni et al., Breed-Dependent Transcriptional Regulation of 5???-Untranslated GR (NR3C1) Exon 1 mRNA Variants in the Liver of Newborn Piglets, PLoS ONE, vol.16, issue.7, p.40432, 2012.
DOI : 10.1371/journal.pone.0040432.s001

J. Kim, G. Song, G. Wu, and F. Bazer, Functional roles of fructose, Proceedings of the National Academy of Sciences, vol.186, issue.3, pp.1619-1647, 2012.
DOI : 10.2741/s184

D. Senger and G. Davis, Angiogenesis, Cold Spring Harbor Perspectives in Biology, vol.3, issue.8, p.5090, 2011.
DOI : 10.1101/cshperspect.a005090

P. Mirtschink, J. Krishnan, F. Grimm, A. Sarre, M. Horl et al., HIF-driven SF3B1 induces KHK-C to enforce fructolysis and heart disease, Nature, vol.121, issue.7557, pp.444-453, 2015.
DOI : 10.1172/JCI44736

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4783869/pdf

E. Mortier, G. Wuytens, I. Leenaerts, F. Hannes, M. Heung et al., Nuclear speckles and nucleoli targeting by PIP2???PDZ domain interactions, The EMBO Journal, vol.12, issue.14, pp.2556-65, 2005.
DOI : 10.1091/mbc.12.2.339

URL : http://emboj.embopress.org/content/embojnl/24/14/2556.full.pdf

L. Drozdowski, T. Clandinin, and A. Thomson, Ontogeny, growth and development of the small intestine: Understanding pediatric gastroenterology, World J Gastroenterol, vol.16, issue.7, pp.787-99, 2010.

F. Gondret, M. Père, S. Tacher, S. Daré, C. Trefeu et al., Spontaneous intra-uterine growth restriction modulates the endocrine status and the developmental expression of genes in porcine fetal and neonatal adipose tissue, General and Comparative Endocrinology, vol.194, pp.208-224, 2013.
DOI : 10.1016/j.ygcen.2013.09.018

URL : https://hal.archives-ouvertes.fr/hal-01210491

C. Désautés, J. Bidanel, and P. Mormède, Genetic Study of Behavioral and Pituitary-Adrenocortical Reactivity in Response to an Environmental Challenge in Pigs, Physiology & Behavior, vol.62, issue.2, pp.337-382, 1997.
DOI : 10.1016/S0031-9384(97)00019-X

M. Hay and P. Mormède, Determination of catecholamines and methoxycatecholamines extraction patterns in pig and rat urine by ion-exchange liquid chromatography with electrochemical detection

F. Smedes, J. Kraak, and H. Poppe, Simple and fast solvent extraction system for selective and quantitative isolation of adrenaline, noradrenaline and dopamine from plasma and urine, Journal of Chromatography B: Biomedical Sciences and Applications, vol.231, issue.1, pp.25-39, 1982.
DOI : 10.1016/S0378-4347(00)80506-X