Comparing the intestinal transcriptome of Meishan and Large White piglets during late fetal development reveals genes involved in glucose and lipid metabolism and immunity as valuable clues of intestinal maturity

BackgroundMaturity of intestinal functions is critical for neonatal health and survival, but comprehensive description of mechanisms underlying intestinal maturation that occur during late gestation still remain poorly characterized. The aim of this study was to investigate biological processes specifically involved in intestinal maturation by comparing fetal jejunal transcriptomes of two representative porcine breeds (Large White, LW; Meishan, MS) with contrasting neonatal vitality and maturity, at two key time points during late gestation (gestational days 90 and 110). MS and LW sows inseminated with mixed semen (from breed LW and MS) gave birth to both purebred and crossbred fetuses. We hypothesized that part of the differences in neonatal maturity between the two breeds results from distinct developmental profiles of the fetal intestine during late gestation. Reciprocal crossed fetuses were used to analyze the effect of parental genome. Transcriptomic data and 23 phenotypic variables known to be associated with maturity trait were integrated using multivariate analysis with expectation of identifying relevant genes-phenotypic variable relationships involved in intestinal maturation.ResultsA moderate maternal genotype effect, but no paternal genotype effect, was observed on offspring intestinal maturation. Four hundred and four differentially expressed probes, corresponding to 274 differentially expressed genes (DEGs), more specifically involved in the maturation process were further studied. In day 110-MS fetuses, Ingenuity® functional enrichment analysis revealed that 46% of DEGs were involved in glucose and lipid metabolism, cell proliferation, vasculogenesis and hormone synthesis compared to day 90-MS fetuses. Expression of genes involved in immune pathways including phagocytosis, inflammation and defense processes was changed in day 110-LW compared to day 90-LW fetuses (corresponding to 13% of DEGs). The transcriptional regulator PPARGC1A was predicted to be an important regulator of differentially expressed genes in MS. Fetal blood fructose level, intestinal lactase activity and villous height were the best predicted phenotypic variables with probes mostly involved in lipid metabolism, carbohydrate metabolism and cellular movement biological pathways.ConclusionsCollectively, our findings indicate that the neonatal maturity of pig intestine may rely on functional development of glucose and lipid metabolisms, immune phagocyte differentiation and inflammatory pathways. This process may partially be governed by PPARGC1A.

[1]  Seymour Geisser,et al.  The Predictive Sample Reuse Method with Applications , 1975 .

[2]  Developmentally regulated tumor necrosis factor-α induced nuclear factor-κB activation in intestinal epithelium , 2007 .

[3]  S. Oddie,et al.  Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants. , 2017, The Cochrane database of systematic reviews.

[4]  R. Derijk,et al.  Glucocorticoid receptor variants: clinical implications , 2002, The Journal of Steroid Biochemistry and Molecular Biology.

[5]  Guoyao Wu,et al.  Temporal proteomic analysis reveals defects in small-intestinal development of porcine fetuses with intrauterine growth restriction. , 2014, The Journal of nutritional biochemistry.

[6]  W. McGuire,et al.  Slow advancement of enteral feed volumes to prevent necrotising enterocolitis in very low birth weight infants. , 2015, The Cochrane database of systematic reviews.

[7]  J. Tyson,et al.  Rapid versus slow rate of advancement of feedings for promoting growth and preventing necrotizing enterocolitis in parenterally fed low-birth-weight infants. , 1998, The Cochrane database of systematic reviews.

[8]  D. Tamandl,et al.  Monocyte Toll-Like Receptor 4 Expression and LPS-Induced Cytokine Production Increase during Gestational Aging , 2005, Pediatric Research.

[9]  Philippe Besse,et al.  Statistical Applications in Genetics and Molecular Biology A Sparse PLS for Variable Selection when Integrating Omics Data , 2011 .

[10]  N. Vain,et al.  Hyperglycaemia in preterm neonates: what to know, what to do. , 2011, Early human development.

[11]  Huan Liu,et al.  Feature Selection for Classification , 1997, Intell. Data Anal..

[12]  O. H. Lowry,et al.  Protein measurement with the Folin phenol reagent. , 1951, The Journal of biological chemistry.

[13]  Mun Chun Chan,et al.  The many roles of PGC-1α in muscle--recent developments. , 2014, Metabolism: clinical and experimental.

[14]  R. Evans,et al.  ERRATUM: PGC-1 promotes insulin resistance in liver through PPAR-α-dependent induction of TRB-3 , 2004, Nature Medicine.

[15]  Guoyao Wu,et al.  Functional roles of fructose , 2012, Proceedings of the National Academy of Sciences.

[16]  I. Koutroubakis,et al.  Multipotent role of platelets in inflammatory bowel diseases: a clinical approach. , 2014, World journal of gastroenterology.

[17]  F. Speleman,et al.  Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes , 2002, Genome Biology.

[18]  M. Patti,et al.  Dual modulation of both lipid oxidation and synthesis by peroxisome proliferator‐activated receptor‐γ coactivator‐1α and ‐1β in cultured myotubes , 2010, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[19]  J. Lunney,et al.  Gene expression profiling in Salmonella Choleraesuis-infected porcine lung using a long oligonucleotide microarray , 2006, Mammalian Genome.

[20]  A. Thomson,et al.  Ontogeny, growth and development of the small intestine: Understanding pediatric gastroenterology. , 2010, World journal of gastroenterology.

[21]  S. Tsai,et al.  Characterization of Conserved and Nonconserved Imprinted Genes in Swine1 , 2009, Biology of reproduction.

[22]  F. Smedes,et al.  Simple and fast solvent extraction system for selective and quantitative isolation of adrenaline, noradrenaline and dopamine from plasma and urine. , 1982, Journal of chromatography.

[23]  M. Sancristobal,et al.  Muscle transcriptomic investigation of late fetal development identifies candidate genes for piglet maturity , 2014, BMC Genomics.

[24]  E. Knol,et al.  Prenatal development as a predisposing factor for perinatal losses in pigs. , 2020, Reproduction (Cambridge, England) Supplement.

[25]  A. Prusa,et al.  Immaturity of infection control in preterm and term newborns is associated with impaired toll-like receptor signaling. , 2007, The Journal of infectious diseases.

[26]  P. Morméde,et al.  Determination of catecholamines and methoxycatecholamines excretion patterns in pig and rat urine by ion-exchange liquid chromatography with electrochemical detection. , 1997, Journal of chromatography. B, Biomedical sciences and applications.

[27]  Marc Montminy,et al.  PGC-1 promotes insulin resistance in liver through PPAR-α-dependent induction of TRB-3 , 2004, Nature Medicine.

[28]  Runsheng Li,et al.  Breed-Dependent Transcriptional Regulation of 5′-Untranslated GR (NR3C1) Exon 1 mRNA Variants in the Liver of Newborn Piglets , 2012, PloS one.

[29]  B. Stoll,et al.  Invited review: the preterm pig as a model in pediatric gastroenterology. , 2013, Journal of animal science.

[30]  S. Cousens,et al.  4 million neonatal deaths: When? Where? Why? , 2005, The Lancet.

[31]  J. Neu Necrotizing Enterocolitis: The Mystery Goes On , 2014, Neonatology.

[32]  J. Thorp,et al.  Intrauterine growth restriction increases morbidity and mortality among premature neonates. , 2004, American journal of obstetrics and gynecology.

[33]  T. Vuocolo,et al.  A gene network switch enhances the oxidative capacity of ovine skeletal muscle during late fetal development , 2010, BMC Genomics.

[34]  A. Mulberg,et al.  Development of the human gastrointestinal tract: twenty years of progress. , 1999, Gastroenterology.

[35]  Alberto D. Pascual-Montano,et al.  GeneCodis3: a non-redundant and modular enrichment analysis tool for functional genomics , 2012, Nucleic Acids Res..

[36]  J. Mei,et al.  The prenatal porcine intestine has low transforming growth factor-beta ligand and receptor density and shows reduced trophic response to enteral diets. , 2009, American journal of physiology. Regulatory, integrative and comparative physiology.

[37]  N. Kretchmer,et al.  Nutritional Adaptation of the Gastrointestinal Tract of the Newborn , 1984, Archives of disease in childhood.

[38]  Zhihua Jiang,et al.  Coordinated miRNA/mRNA Expression Profiles for Understanding Breed-Specific Metabolic Characters of Liver between Erhualian and Large White Pigs , 2012, PloS one.

[39]  W. Walker,et al.  Glucocorticoid responsiveness in developing human intestine: possible role in prevention of necrotizing enterocolitis. , 2005, American journal of physiology. Gastrointestinal and liver physiology.

[40]  B. Stoll,et al.  Dual purpose use of preterm piglets as a model of pediatric GI disease. , 2014, Veterinary immunology and immunopathology.

[41]  I. Le Huërou-Luron,et al.  Intrauterine growth restriction modifies the developmental pattern of intestinal structure, transcriptomic profile, and bacterial colonization in neonatal pigs. , 2010, The Journal of nutrition.

[42]  D. Val-Laillet,et al.  Critical review evaluating the pig as a model for human nutritional physiology , 2016, Nutrition Research Reviews.

[43]  Andreas Krämer,et al.  Causal analysis approaches in Ingenuity Pathway Analysis , 2013, Bioinform..

[44]  Ignacio González,et al.  integrOmics: an R package to unravel relationships between two omics datasets , 2009, Bioinform..

[45]  M. Père,et al.  Spontaneous intra-uterine growth restriction modulates the endocrine status and the developmental expression of genes in porcine fetal and neonatal adipose tissue. , 2013, General and comparative endocrinology.

[46]  E. Knol,et al.  Fetal development in the pig in relation to genetic merit for piglet survival. , 2002, Journal of animal science.

[47]  M. Père Materno-foetal exchanges and utilisation of nutrients by the foetus: comparison between species. , 2003, Reproduction, nutrition, development.

[48]  J. Miles,et al.  Placental accommodations for transport and metabolism during intra-uterine crowding in pigs , 2014, Journal of Animal Science and Biotechnology.

[49]  D. Ménard,et al.  Developmental aspects of lipid and lipoprotein synthesis and secretion in human gut , 2000, Microscopy research and technique.

[50]  C David,et al.  Estimation of genetic trends from 1977 to 1998 of body composition and physiological state of Large White pigs at birth. , 2007, Animal : an international journal of animal bioscience.

[51]  Joy E. Lawn,et al.  Levels and Causes of Mortality under Age Five Years , 2016 .

[52]  S. Rautava,et al.  TGF-&bgr;2 Induces Maturation of Immature Human Intestinal Epithelial Cells and Inhibits Inflammatory Cytokine Responses Induced Via the NF-&kgr;B Pathway , 2012, Journal of pediatric gastroenterology and nutrition.

[53]  N. Ambalavanan,et al.  TGF-β2 suppresses macrophage cytokine production and mucosal inflammatory responses in the developing intestine. , 2011, Gastroenterology.

[54]  J. L. Dividich,et al.  Early Weaning Stimulates Intestinal Brush Border Enzyme Activities in Piglets, Mainly at the Posttranscriptional Level , 2005, Journal of pediatric gastroenterology and nutrition.

[55]  P. Zimmermann,et al.  Nuclear speckles and nucleoli targeting by PIP2–PDZ domain interactions , 2005, The EMBO journal.

[56]  J. Neu,et al.  Decoding the enigma of necrotizing enterocolitis in premature infants. , 2014, Pathophysiology : the official journal of the International Society for Pathophysiology.

[57]  R. Buddington,et al.  Prenatal Development of Gastrointestinal Function in the Pig and the Effects of Fetal Esophageal Obstruction , 2002, Pediatric Research.

[58]  G. Randall,et al.  Tissue glycogen and blood glucose and fructose levels in the pig fetus during the second half of gestation. , 1976, Biology of the neonate.

[59]  J. Orenstein,et al.  Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. , 2005, The Journal of clinical investigation.

[60]  J. Wan,et al.  Premature Delivery Reduces Intestinal Cytoskeleton, Metabolism, and Stress Response Proteins in Newborn Formula-Fed Pigs , 2013, Journal of pediatric gastroenterology and nutrition.

[61]  J. Ule,et al.  HIF-driven SF3B1 induces KHK-C to enforce fructolysis and heart disease , 2015, Nature.

[62]  A. Fowden,et al.  How does the foetal gastrointestinal tract develop in preparation for enteral nutrition after birth , 2000 .

[63]  M. Stone Cross‐Validatory Choice and Assessment of Statistical Predictions , 1976 .

[64]  A. Maheshwari Immunologic and Hematological Abnormalities in Necrotizing Enterocolitis. , 2015, Clinics in perinatology.

[65]  L Canario,et al.  Between-breed variability of stillbirth and its relationship with sow and piglet characteristics. , 2006, Journal of animal science.

[66]  Reduced TNF‐α response in preterm neonates is associated with impaired nonclassic monocyte function , 2016, Journal of leukocyte biology.

[67]  H. Nauwynck,et al.  Scavenger receptor CD163, a Jack-of-all-trades and potential target for cell-directed therapy. , 2010, Molecular immunology.

[68]  J. Bidanel,et al.  Genetic Study of Behavioral and Pituitary-Adrenocortical Reactivity in Response to an Environmental Challenge in Pigs , 1997, Physiology & Behavior.

[69]  A. Stevens,et al.  The metabolomics of necrotising enterocolitis in preterm babies: an exploratory study , 2016, The journal of maternal-fetal & neonatal medicine : the official journal of the European Association of Perinatal Medicine, the Federation of Asia and Oceania Perinatal Societies, the International Society of Perinatal Obstetricians.

[70]  D. Sabatini,et al.  mTOR: from growth signal integration to cancer, diabetes and ageing , 2010, Nature Reviews Molecular Cell Biology.