. Obesity causes substantial social, economic and health burdens. The rate of obesity is escalating disproportionately in children (infants to young adults). This rapid increase is unlikely to be due to environment or genetics alone. Accumulating evidence from our laboratory and others suggests that adult metabolic diseases originate in utero, and likely occur through the reprogramming of gene expression via epigenetic changes in chromatin structure (an altered """"""""histone code""""""""). Of interest, we have observed in a rodent transgenerational model of intrauterine growth restriction (IUGR) that a diet supplemented with essential nutrients, yet unaltered in its caloric content, prevents adult metabolic disease and is associated with abrogation of reprogrammed gene expression. However, although such established models in rodents demonstrate that fetal alterations in the histone code are involved in the persistence and conveyance of the altered postnatal phenotype, little is known about the effects of maternal diet and resultant obesity on primate fetal biology. We hypothesized that a high fat diet in non-human primates would induce changes in hepatic chromatin structure resulting in altered expression of fetal genes critical to the development of childhood and adult obesity. Based on our preliminary data, the focus of this proposal is to apply developed high throughput technology (comparative epigenomics and metabolomics) to decipher the primate epigenome and metabolome in the obese maternal environment and then measure the impact of supplementation on the differentially altered epigenome and resultant disease. The novel innovation and significance resides within its potential to provide (1) an expanded understanding of the mechanism through which a maternal high fat diet reprograms primate gene expression and (2) a simple intervention (essential nutrient supplementation with neither diet nor behavioral modification) with tremendous potential impact given the current obesity epidemic and the lack of efficacious therapeutics.

Agency
National Institute of Health (NIH)
Institute
Office of The Director, National Institutes of Health (OD)
Type
NIH Director’s New Innovator Awards (DP2)
Project #
3DP2OD001500-01S2
Application #
8289953
Study Section
Special Emphasis Panel (ZGM1-NDIA-G (02))
Program Officer
Basavappa, Ravi
Project Start
2007-09-30
Project End
2012-08-31
Budget Start
2007-09-30
Budget End
2012-08-31
Support Year
1
Fiscal Year
2011
Total Cost
$3,916
Indirect Cost
Name
Baylor College of Medicine
Department
Obstetrics & Gynecology
Type
Schools of Medicine
DUNS #
051113330
City
Houston
State
TX
Country
United States
Zip Code
77030
Pace, Ryan M; Prince, Amanda L; Ma, Jun et al. (2018) Modulations in the offspring gut microbiome are refractory to postnatal synbiotic supplementation among juvenile primates. BMC Microbiol 18:28
Chu, Derrick M; Ma, Jun; Prince, Amanda L et al. (2017) Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat Med 23:314-326
O'Neil, Derek S; Stewart, Christopher J; Chu, Derrick M et al. (2017) Conditional postnatal deletion of the neonatal murine hepatic circadian gene, Npas2, alters the gut microbiome following restricted feeding. Am J Obstet Gynecol 217:218.e1-218.e15
Chu, Derrick M; Antony, Kathleen M; Ma, Jun et al. (2016) The early infant gut microbiome varies in association with a maternal high-fat diet. Genome Med 8:77
Harris, R Alan; Alcott, Callison E; Sullivan, Elinor L et al. (2016) Genomic Variants Associated with Resistance to High Fat Diet Induced Obesity in a Primate Model. Sci Rep 6:36123
Prince, Amanda L; Ma, Jun; Kannan, Paranthaman S et al. (2016) The placental membrane microbiome is altered among subjects with spontaneous preterm birth with and without chorioamnionitis. Am J Obstet Gynecol 214:627.e1-627.e16
Kahr, Maike K; Suter, Melissa A; Ballas, Jerasimos et al. (2016) Geospatial analysis of food environment demonstrates associations with gestational diabetes. Am J Obstet Gynecol 214:110.e1-9
Pew, Braden K; Harris, R Alan; Sbrana, Elena et al. (2016) Structural and transcriptomic response to antenatal corticosteroids in an Erk3-null mouse model of respiratory distress. Am J Obstet Gynecol 215:384.e1-384.e89
Gonzalez-Rodriguez, Pablo; Cantu, Jessica; O'Neil, Derek et al. (2016) Alterations in expression of imprinted genes from the H19/IGF2 loci in a multigenerational model of intrauterine growth restriction (IUGR). Am J Obstet Gynecol 214:625.e1-625.e11
Seferovic, Maxim D; Goodspeed, Danielle M; Chu, Derrick M et al. (2015) Heritable IUGR and adult metabolic syndrome are reversible and associated with alterations in the metabolome following dietary supplementation of 1-carbon intermediates. FASEB J 29:2640-52

Showing the most recent 10 out of 47 publications