Skip to main content

Advertisement

Log in

Epigenetics of Childhood Obesity

  • Obesity (S Armstrong and A Patel, Section Editors)
  • Published:
Current Pediatrics Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

We sought to describe how epigenetics, the study of potentially heritable changes in gene expression that do not involve changes in the underlying DNA sequence, has promise in the development of effective preventive interventions for childhood obesity.

Recent Findings

Three main mechanisms cause epigenetic changes: DNA methylation, histone and chromatin modification, and post-translational regulation, including that by microRNAs (miRNAs). These mechanisms may be altered by a variety of exposures, and evidence suggests they act at multiple time points prior to conception, during pregnancy, and during infancy and childhood. Epigenetic changes, particularly DNA methylation, have been associated with birth weight, early childhood, and teenage obesity. Each epigenetic mechanism raises the possibility of therapeutics. DNA methylation may be affected by maternal and paternal diet, exercise, and supplements. Histone and chromatin modification can be altered by histone deacetylase inhibitors and other small molecules. miRNAs are already being studied in cancer and type 2 diabetes.

Summary

Although epigenetics is in its infancy, it could provide an important link between maternal exposures and childhood metabolic outcomes.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance

  1. Whitaker RC, et al. Predicting obesity in young adulthood from childhood and parental obesity. N Engl J Med. 1997;337(13):869–73.

    Article  CAS  PubMed  Google Scholar 

  2. Llewellyn CH, et al. Eating rate is a heritable phenotype related to weight in children. Am J Clin Nutr. 2008;88(6):1560–6.

    Article  CAS  PubMed  Google Scholar 

  3. Han TS, et al. Contributions of maternal and paternal adiposity and smoking to adult offspring adiposity and cardiovascular risk: the Midspan Family Study. BMJ Open. 2015;5(11):e007682.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Waterland RA, Jirtle RL. Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol Cell Biol. 2003;23(15):5293–300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Painter RC, Roseboom TJ, Bleker OP. Prenatal exposure to the Dutch famine and disease in later life: an overview. Reprod Toxicol. 2005;20(3):345–52.

    Article  CAS  PubMed  Google Scholar 

  6. Jirtle RL, Skinner MK. Environmental epigenomics and disease susceptibility. Nat Rev Genet. 2007;8(4):253–62.

    Article  CAS  PubMed  Google Scholar 

  7. Heijmans BT, et al. Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc Natl Acad Sci. 2008;105(44):17046–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kappil M, Wright RO, Sanders AP. Developmental origins of common disease: epigenetic contributions to obesity. Annu Rev Genomics hum Genet. 2016;17(1):177–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Murrell A. An association between variants in the IGF2 gene and Beckwith-Wiedemann syndrome: interaction between genotype and epigenotype. Hum Mol Genet. 2003;13(2):247–55.

    Article  PubMed  Google Scholar 

  10. Waterland RA, Michels KB. Epigenetic epidemiology of the developmental origins hypothesis. Annu Rev Nutr. 2007;27(1):363–88.

    Article  CAS  PubMed  Google Scholar 

  11. Ferraro ZM, et al. Excessive gestational weight gain predicts large for gestational age neonates independent of maternal body mass index. J Matern Fetal Neonatal Med. 2011;25(5):538–42.

    Article  PubMed  Google Scholar 

  12. Linabery AM, et al. Stronger influence of maternal than paternal obesity on infant and early childhood body mass index: the Fels Longitudinal Study. Pediatric Obesity. 2012;8(3):159–69.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Starling AP, et al. Associations of maternal BMI and gestational weight gain with neonatal adiposity in the Healthy Start study. Am J Clin Nutr. 2014;101(2):302–9.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Krikke GG, et al. Vitamin B12and folate status in early pregnancy and cardiometabolic risk factors in the offspring at age 5-6 years: findings from the ABCD multi-ethnic birth cohort. BJOG Int J Obstet Gynaecol. 2015;123(3):384–92.

    Article  Google Scholar 

  15. Richmond RC, et al. Using genetic variation to explore the causal effect of maternal pregnancy adiposity on future offspring adiposity: a Mendelian Randomisation Study. PLoS med. 2017;14(1):e1002221.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Vidal AC, et al. Associations between antibiotic exposure during pregnancy, birth weight and aberrant methylation at imprinted genes among offspring. Int J Obes. 2013;37(7):907–13.

    Article  CAS  Google Scholar 

  17. Song Y, et al. Transgenerational impaired male fertility with an Igf2 epigenetic defect in the rat are induced by the endocrine disruptor p,p'-DDE. Hum Reprod. 2014;29(11):2512–21.

    Article  PubMed  Google Scholar 

  18. Ma J, et al. Ancestral TCDD exposure promotes epigenetic transgenerational inheritance of imprinted gene Igf2: methylation status and DNMTs. Toxicol Appl Pharmacol. 2015;289(2):193–202.

    Article  CAS  PubMed  Google Scholar 

  19. Dhasarathy A, Roemmich JN, Claycombe KJ. Influence of maternal obesity, diet and exercise on epigenetic regulation of adipocytes. Mol Aspects Med. 2016.

  20. Soubry A, et al. Obesity-related DNA methylation at imprinted genes in human sperm: results from the TIEGER study. Clin Epigenetics. 2016;8(1).

  21. Agha G, et al. Birth weight-for-gestational age is associated with DNA methylation at birth and in childhood. Clin Epigenet. 2016;8(1).

  22. Kadakia R, et al. Maternal pre-pregnancy BMI downregulates neonatal cord blood LEP methylation. Pediatr Obes. 2016.

  23. McCullough LE, et al. Associations between prenatal physical activity, birth weight, and DNA methylation at genomically imprinted domains in a multiethnic newborn cohort. Epigenetics. 2015;10(7):597–606.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Huang RC, et al. Genome-wide methylation analysis identifies differentially methylated CpG loci associated with severe obesity in childhood. Epigenetics. 2015;10(11):995–1005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Soubry A, et al. Newborns of obese parents have altered DNA methylation patterns at imprinted genes. Int J Obes 2015;39(4): 650–7.

  26. Wang S, et al. HIF3A DNA methylation is associated with childhood obesity and ALT. PLoS One. 2015;10(12):e0145944.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Engel SM, et al. Neonatal genome-wide methylation patterns in relation to birth weight in the Norwegian Mother and Child Cohort. Am J Epidemiol. 2014;179(7):834–42.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Haworth KE, et al. Methylation of the FGFR2 gene is associated with high birth weight centile in humans. Epigenomics. 2014;6(5):477–91.

    Article  CAS  PubMed  Google Scholar 

  29. Turan N, et al. DNA methylation differences at growth related genes correlate with birth weight: a molecular signature linked to developmental origins of adult disease? BMC Med Genomics. 2012;5(1).

  30. Hoyo C, et al. Association of cord blood methylation fractions at imprinted insulin-like growth factor 2 (IGF2), plasma IGF2, and birth weight. Cancer Causes Control. 2012;23(4):635–45.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Suter MA, et al. A maternal high-fat diet modulates fetal SIRT1 histone and protein deacetylase activity in nonhuman primates. FASEB J. 2012;26(12):5106–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Prince CS, Maloyan A, Myatt L. Maternal obesity alters brain derived neurotrophic factor (BDNF) signaling in the placenta in a sexually dimorphic manner. Placenta. 2017;49:55–63.

    Article  CAS  PubMed  Google Scholar 

  33. Godfrey KM, et al. Epigenetic gene promoter methylation at birth is associated with child’s later adiposity. Diabetes. 2011;60(5):1528–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Grundberg E, et al. Global analysis of DNA methylation variation in adipose tissue from twins reveals links to disease-associated variants in distal regulatory elements. Am J Hum Genet. 2013;93(6):1158.

    Article  CAS  PubMed Central  Google Scholar 

  35. Morales E, et al. DNA methylation signatures in cord blood associated with maternal gestational weight gain: results from the ALSPAC cohort. BMC Research Notes. 2014;7(1):278.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Sharp GC, et al. Maternal pre-pregnancy BMI and gestational weight gain, offspring DNA methylation and later offspring adiposity: findings from the Avon Longitudinal Study of Parents and Children. Int J Epidemiol. 2015;44(4):1288–304.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Ding X, et al. Genome-wide screen of DNA methylation identifies novel markers in childhood obesity. Gene. 2015;566(1):74–83.

    Article  CAS  PubMed  Google Scholar 

  38. Bastie CC, et al. Integrative metabolic regulation of peripheral tissue fatty acid oxidation by the SRC kinase family member Fyn. Cell Metab. 2007;5(5):371–81.

    Article  CAS  PubMed  Google Scholar 

  39. Tse MCL, et al. Fyn regulates adipogenesis by promoting PIKE-A/STAT5a interaction. Mol Cell Biol. 2013;33(9):1797–808.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. DeChiara TM, Efstratiadis A, Robertsen EJ. A growth-deficiency phenotype in heterozygous mice carrying an insulin-like growth factor II gene disrupted by targeting. Nature. 1990;345(6270):78–80.

    Article  CAS  PubMed  Google Scholar 

  41. Vu TH, et al. Symmetric and asymmetric DNA methylation in the human IGF2–H19 imprinted region. Genomics. 2000;64(2):132–43.

    Article  CAS  PubMed  Google Scholar 

  42. Soubry A, et al. The effects of depression and use of antidepressive medicines during pregnancy on the methylation status of the IGF2 imprinted control regions in the offspring. Clin Epigenetics. 2011;3(1):2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Soubry A, et al, Paternal obesity is associated with IGF2hypomethylation in newborns: results from a Newborn Epigenetics Study (NEST) cohort. BMC Med. 2013;11(1).

  44. St-Pierre J, et al. IGF2 DNA methylation is a modulator of newborn’s fetal growth and development. Epigenetics. 2012;7(10):1125–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. • Perkins E, et al. Insulin-like growth factor 2/H19 methylation at birth and risk of overweight and obesity in children. J Pediatr. 2012;161(1):31–9. This study examined IGF2 imprinting region DNA methylation in relation to body weight at age 1 year, and suggested that regulation of imprinted genes could have an important influence on childhood obesity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hernández-Valero MA, et al. Interplay between polymorphisms and methylation in the H19/IGF2 gene region may contribute to obesity in Mexican-American children. J Dev Orig Health Dis. 2013;4(06):499–506.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Broholm C, et al. Human adipogenesis is associated with genome-wide DNA methylation and gene-expression changes. Epigenomics. 2016;8(12):1601–17.

    Article  CAS  PubMed  Google Scholar 

  48. Zhang M, et al. Leptin and leptin-to-adiponectin ratio predict adiposity gain in nonobese children over a six-year period. Childhood Obes. 2017.

  49. Broholm C, et al. Epigenetic programming of adipose-derived stem cells in low birthweight individuals. Diabetologia. 2016;59(12):2664–73.

    Article  PubMed  Google Scholar 

  50. Josefson JL, et al. Maternal leptin predicts adiposity of the neonate. Hormone Research in Paediatrics. 2014;81(1):13–9.

    Article  CAS  PubMed  Google Scholar 

  51. Houde A-A, et al. Leptin and adiponectin DNA methylation levels in adipose tissues and blood cells are associated with BMI, waist girth and LDL-cholesterol levels in severely obese men and women. BMC Med Genet. 2015;16(1).

  52. Fandy TE, Carraway H, Gore SD. DNA demethylating agents and histone deacetylase inhibitors in hematologic malignancies. The Cancer Journal. 2007;13(1):40–8.

    Article  CAS  PubMed  Google Scholar 

  53. Vendetti FP, Rudin CM. Epigenetic therapy in non-small-cell lung cancer: targeting DNA methyltransferases and histone deacetylases. Expert Opin Biol Ther. 2013;13(9):1273–85.

    Article  CAS  PubMed  Google Scholar 

  54. Zhou D, et al. High fat diet and exercise lead to a disrupted and pathogenic DNA methylome in mouse liver. Epigenetics. 2016;12(1):55–69.

    Article  PubMed  Google Scholar 

  55. Laker RC, et al. Exercise prevents maternal high-fat diet–induced hypermethylation of the Pgc-1αGene and age-dependent metabolic dysfunction in the offspring. Diabetes. 2014;63(5):1605–11.

    Article  CAS  PubMed  Google Scholar 

  56. Wasinski F, et al. Exercise during pregnancy protects adult mouse offspring from diet-induced obesity. Nutr Metab. 2015;12(1).

  57. Milagro FI, et al. A dual epigenomic approach for the search of obesity biomarkers: DNA methylation in relation to diet-induced weight loss. FASEB j. 2011;25(4):1378–89.

    Article  CAS  PubMed  Google Scholar 

  58. Rönn T, et al. A six months exercise intervention influences the genome-wide DNA methylation pattern in human adipose tissue. PLoS Genet. 2013;9(6):e1003572.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Azzi S, et al. Degree of methylation of ZAC1 (PLAGL1) is associated with prenatal and post-natal growth in healthy infants of the EDEN mother child cohort. Epigenetics. 2014;9(3):338–45.

    Article  CAS  PubMed  Google Scholar 

  60. Hopkins SA, et al. Exercise training in pregnancy reduces offspring size without changes in maternal insulin sensitivity. Obstetrical & Gynecological Survey. 2010;65(9):565–6.

    Article  Google Scholar 

  61. Hopkins SA, et al. Effects of exercise training on maternal hormonal changes in pregnancy. Clin Endocrinol. 2011;74(4):495–500.

    Article  Google Scholar 

  62. Crider KS, et al. Genomic DNA methylation changes in response to folic acid supplementation in a population-based intervention study among women of reproductive age. PLoS One. 2011;6(12):e28144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Dominguez-Salas P, et al. Maternal nutrition at conception modulates DNA methylation of human metastable epialleles. Nat Commun. 2014;5

  64. Lee HS, et al. Dietary supplementation with polyunsaturated fatty acid during pregnancy modulates DNA methylation at IGF2/H19 imprinted genes and growth of infants. Physiol Genomics. 2014;46(23):851–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Boqué N, et al. Prevention of diet-induced obesity by apple polyphenols in Wistar rats through regulation of adipocyte gene expression and DNA methylation patterns. Mol Nutr Food res. 2013;57(8):1473–8.

    Article  PubMed  Google Scholar 

  66. Viswanathan M, et al. Folic acid supplementation for the prevention of neural tube defects: an updated evidence report and systematic review for the US Preventive Services Task Force. JAMA. 2017;317(2):190–203.

    Article  PubMed  Google Scholar 

  67. Bricambert J, et al. Impaired histone deacetylases 5 and 6 expression mimics the effects of obesity and hypoxia on adipocyte function. Molecular Metabolism. 2016;5(12):1200–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Ferrari A, et al. Attenuation of diet-induced obesity and induction of white fat browning with a chemical inhibitor of histone deacetylases. Int J Obes. 2016;41(2):289–98.

    Article  Google Scholar 

  69. Remsberg JR, et al. Deletion of histone deacetylase 3 in adult beta cells improves glucose tolerance via increased insulin secretion. Molecular Metabolism. 2017;6(1):30–7.

    Article  CAS  PubMed  Google Scholar 

  70. Arguelles AO, et al. Are epigenetic drugs for diabetes and obesity at our door step? Drug Discov Today. 2016;21(3):499–509.

    Article  PubMed  Google Scholar 

  71. Lee M, et al. The gender association of the SIRT1 rs7895833 polymorphism with pediatric obesity: a 3-year panel study. J Nutrigenet Nutrigenomics. 2016;265–275.

  72. Ferrante SC, et al. Adipocyte-derived exosomal miRNAs: a novel mechanism for obesity-related disease. Pediatr res. 2014;77(3):447–54.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Hubal MJ, et al. Circulating adipocyte-derived exosomal MicroRNAs associated with decreased insulin resistance after gastric bypass. Obesity. 2016;25(1):102–10.

    Article  PubMed  Google Scholar 

  74. Li J, et al. Small non-coding RNAs transfer through mammalian placenta and directly regulate fetal gene expression. Protein & Cell. 2015;6(6):391–6.

    Article  Google Scholar 

  75. Murashov AK, et al. Paternal long-term exercise programs offspring for low energy expenditure and increased risk for obesity in mice. FASEB j. 2015;30(2):775–84.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Sanders AP, et al. Altered miRNA expression in the cervix during pregnancy associated with lead and mercury exposure. Epigenomics. 2015;7(6):885–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Rosas-Vargas H, Martínez-Ezquerro JD, Bienvenu T. Brain-derived neurotrophic factor, food intake regulation, and obesity. Arch med res. 2011;42(6):482–94.

    Article  CAS  PubMed  Google Scholar 

  78. Gardner KR, Sapienza C, Fisher JO. Genetic and epigenetic associations to obesity-related appetite phenotypes among African-American children. Pediatric Obesity. 2015;10(6):476–82.

    Article  CAS  PubMed  Google Scholar 

  79. Kurylowicz A, et al. SIRT1 and SIRT7 expression in adipose tissues of obese and normal-weight individuals is regulated by microRNAs but not by methylation status. Int J Obes. 2016;40(11):1635–42.

    Article  CAS  Google Scholar 

  80. Robb, T., G. Reid, and C. Blenkiron, Exploiting microRNAs as cancer therapeutics. Targeted Oncology, 2017.

    Google Scholar 

  81. Link JC, et al. Diet, gonadal sex, and sex chromosome complement influence white adipose tissue miRNA expression. BMC Genomics. 2017;18(1).

  82. Ortega FJ, et al. Profiling of circulating microRNAs reveals common microRNAs linked to type 2 diabetes that change with insulin sensitization. Diabetes Care. 2014;37(5):1375–83.

    Article  CAS  PubMed  Google Scholar 

  83. • Simpkin AJ, et al. Longitudinal analysis of DNA methylation associated with birth weight and gestational age. Human Mol Genet. 2015. This is a study from the Avon Longitudinal Study of Parents and Children (ALSPAC), a large longitudinal cohort. This study assessed the durability of DNA methylation changes at ages 7 and 17 years, one of the first studies to look at DNA methylation in the teenage years. Persistence of DNA methylation is an essential question in the field of epigenetics.

  84. Loucks EB, et al. Epigenetic mediators between childhood socioeconomic disadvantage and mid-life body mass index. Psychosom med. 2016;78(9):1053–65.

    Article  Google Scholar 

  85. Murphy SK, Huang Z, Hoyo C. Differentially methylated regions of imprinted genes in prenatal, perinatal and postnatal human tissues. PLoS One. 2012;7(7):e40924.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Corsi DJ, et al. Is there a greater maternal than paternal influence on offspring adiposity in India? Arch Dis Child. 2015;100(10):973–9.

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Cynthia A. Moylan.

Ethics declarations

Conflict of Interest

Kara Wegermann and Cynthia A. Moylan each declare no potential conflicts of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

This article is part of the Topical Collection on Obesity

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wegermann, K., Moylan, C.A. Epigenetics of Childhood Obesity. Curr Pediatr Rep 5, 111–117 (2017). https://doi.org/10.1007/s40124-017-0133-8

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40124-017-0133-8

Keywords

Navigation