Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Endocrine-disrupting chemicals and fatty liver disease

Key Points

  • Nonalcoholic fatty liver disease (NAFLD) is a growing epidemic in countries that consume a Western diet, and it can lead to irreversible cirrhosis and hepatocellular carcinoma

  • Exposure to endocrine-disrupting chemicals (EDCs) in early life could represent a 'new' risk factor for the development of NAFLD later in life

  • The mechanism of action of EDCs involves both the modulation of nuclear hormone receptor function via co-regulator proteins and the alteration of the epigenome (that is, DNA methylation and histone modification)

  • Animal model studies suggest causality between early-life exposure to certain EDCs and NAFLD presentation later in life

  • Studies are needed to define whether there is a causal relationship between EDC exposure and development of NAFLD in humans, as well as to develop new prevention and treatment regimes

Abstract

A growing epidemic of nonalcoholic fatty liver disease (NAFLD) is paralleling the increase in the incidence of obesity and diabetes mellitus in countries that consume a Western diet. As NAFLD can lead to life-threatening conditions such as cirrhosis and hepatocellular carcinoma, an understanding of the factors that trigger its development and pathological progression is needed. Although by definition this disease is not associated with alcohol consumption, exposure to environmental agents that have been linked to other diseases might have a role in the development of NAFLD. Here, we focus on one class of these agents, endocrine-disrupting chemicals (EDCs), and their potential to influence the initiation and progression of a cascade of pathological conditions associated with hepatic steatosis (fatty liver). Experimental studies have revealed several potential mechanisms by which EDC exposure might contribute to disease pathogenesis, including the modulation of nuclear hormone receptor function and the alteration of the epigenome. However, many questions remain to be addressed about the causal link between acute and chronic EDC exposure and the development of NAFLD in humans. Future studies that address these questions hold promise not only for understanding the linkage between EDC exposure and liver disease but also for elucidating the molecular mechanisms that underpin NAFLD, which in turn could facilitate the development of new prevention and treatment opportunities.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The pathophysiology of nonalcoholic fatty liver disease progression.
Figure 2: Altered hepatic metabolic pathways that lead to nonalcoholic fatty liver disease.
Figure 3: Nuclear hormone receptor-mediated effects of endocrine- disrupting chemicals on the development of steatosis.
Figure 4: The potential genomic mechanism of action of endocrine-disrupting chemicals.
Figure 5: The epigenomic action of 'writers' of DNA or histone methylation.
Figure 6: Early-life exposure to endocrine-disrupting chemicals triggers the development of nonalcoholic fatty liver disease.
Figure 7: Exposure to endocrine-disrupting chemicals and the risk of nonalcoholic fatty liver disease across the life course.

Similar content being viewed by others

References

  1. Lim, J. S., Mietus-Snyder, M., Valente, A., Schwarz, J. M. & Lustig, R. H. The role of fructose in the pathogenesis of NAFLD and the metabolic syndrome. Nat. Rev. Gastroenterol. Hepatol. 7, 251–264 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Wesolowski, S. R., Kasmi, K. C., Jonscher, K. R. & Friedman, J. E. Developmental origins of NAFLD: a womb with a clue. Nat. Rev. Gastroenterol. Hepatol. 14, 81–96 (2016). This review presents the hypothesis that early-life environmental signals, such as distinct nutritional signals, might predispose an individual to the development of NAFLD in later life.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Wree, A., Broderick, L., Canbay, A., Hoffman, H. M. & Feldstein, A. E. From NAFLD to NASH to cirrhosis — new insights into disease mechanisms. Nat. Rev. Gastroenterol. Hepatol. 10, 627–636 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Michelotti, G. A., Machado, M. V. & Diehl, A. M. NAFLD, NASH and liver cancer. Nat. Rev. Gastroenterol. Hepatol. 10, 656–665 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Hardy, T., Oakley, F., Anstee, Q. M. & Day, C. P. Nonalcoholic fatty liver disease: pathogenesis and disease spectrum. Annu. Rev. Pathol. 11, 451–496 (2016). This review describes the liver pathophysiology that underlies NAFLD.

    Article  CAS  PubMed  Google Scholar 

  6. White, D. L., Kanwal, F. & El-Serag, H. B. Association between nonalcoholic fatty liver disease and risk for hepatocellular cancer, based on systematic review. Clin. Gastroenterol. Hepatol. 10, 1342–1359.e2 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Loomba, R. & Sanyal, A. J. The global NAFLD epidemic. Nat. Rev. Gastroenterol. Hepatol. 10, 686–690 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Ruhl, C. E. & Everhart, J. E. Fatty liver indices in the multiethnic United States National Health and Nutrition Examination Survey. Aliment. Pharmacol. Ther. 41, 65–76 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Ogden, C. L., Carroll, M. D., Kit, B. K. & Flegal, K. M. Prevalence of childhood and adult obesity in the United States, 2011–2012. JAMA 311, 806–814 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Satapathy, S. K. & Sanyal, A. J. Epidemiology and natural history of nonalcoholic fatty liver disease. Semin. Liver Dis. 35, 221–235 (2015).

    Article  PubMed  Google Scholar 

  11. Rinella, M. E. Nonalcoholic fatty liver disease: a systematic review. JAMA 313, 2263–2273 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Heindel, J. J. et al. Metabolism disrupting chemicals and metabolic disorders. Reprod. Toxicol. 68, 3–33 (2016). This timely review describes EDCs that affect metabolic 'set points' and suggests that EDCs should now be called metabolism-disrupting chemicals.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Gore, A. C. et al. EDC-2: The Endocrine Society's second scientific statement on endocrine-disrupting chemicals. Endocr. Rev. 36, E1–E150 (2015). This is an authoritative review of the collective literature on EDCs and their effects on various target organs.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Calafat, A. M. et al. Urinary concentrations of bisphenol A and 4-nonylphenol in a human reference population. Environ. Health Perspect. 113, 391–395 (2005).

    Article  CAS  PubMed  Google Scholar 

  15. Desmet, V. J. in The Liver: Biology and Pathobiology 3rd edn (eds Arias, I. M. et al.) 425–476 (Raven Press, 1994).

    Google Scholar 

  16. Hales, C. N. & Barker, D. J. Type 2 (non-insulin-dependent) diabetes mellitus: the thrifty phenotype hypothesis. Diabetologia 35, 595–601 (1992).

    Article  CAS  PubMed  Google Scholar 

  17. Ravelli, A. C. et al. Glucose tolerance in adults after prenatal exposure to famine. Lancet 351, 173–177 (1998).

    Article  CAS  PubMed  Google Scholar 

  18. Roseboom, T. J. et al. Plasma lipid profiles in adults after prenatal exposure to the Dutch famine. Am. J. Clin. Nutr. 72, 1101–1106 (2000).

    Article  CAS  PubMed  Google Scholar 

  19. Wang, N. et al. Exposure to famine in early life and nonalcoholic fatty liver disease in adulthood. J. Clin. Endocrinol. Metab. 101, 2218–2225 (2016). This paper highlights important human data that were collected during the Great Chinese Famine and shows that a lack of adequate nutrition in early life seems to predispose individuals to the development of NAFLD in later life.

    Article  CAS  PubMed  Google Scholar 

  20. Erslev, A. J. in The Liver: Biology and Pathobiology (eds Arias, I. M. et al.) 1227–1234 (Raven Press, 1994).

    Google Scholar 

  21. Kuntz, E. & Kuntz, H. D. in Hepatology — Textbook and Atlas (eds Kuntz, E. & Kuntz, H. D.) 35–76 (Springer, 2008).

    Book  Google Scholar 

  22. Saponaro, C., Gaggini, M. & Gastaldelli, A. Nonalcoholic fatty liver disease and type 2 diabetes: common pathophysiologic mechanisms. Curr. Diab. Rep. 15, 607 (2015).

    Article  PubMed  CAS  Google Scholar 

  23. Jou, J., Choi, S. S. & Diehl, A. M. Mechanisms of disease progression in nonalcoholic fatty liver disease. Semin. Liver Dis. 28, 370–379 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. De Taeye, B. M. et al. Macrophage TNF-α contributes to insulin resistance and hepatic steatosis in diet-induced obesity. Am. J. Physiol. Endocrinol. Metab. 293, E713–E725 (2007).

    Article  CAS  PubMed  Google Scholar 

  25. Malaguarnera, L., Madeddu, R., Palio, E., Arena, N. & Malaguarnera, M. Heme oxygenase-1 levels and oxidative stress-related parameters in non-alcoholic fatty liver disease patients. J. Hepatol. 42, 585–591 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Papandreou, D. & Andreou, E. Role of diet on non-alcoholic fatty liver disease: an updated narrative review. World J. Hepatol. 7, 575–582 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Reeves, H. L., Zaki, M. Y. & Day, C. P. Hepatocellular carcinoma in obesity, type 2 diabetes, and NAFLD. Dig. Dis. Sci. 61, 1234–1245 (2016).

    Article  CAS  PubMed  Google Scholar 

  28. Hooper, A. J., Adams, L. A. & Burnett, J. R. Genetic determinants of hepatic steatosis in man. J. Lipid Res. 52, 593–617 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ouyang, X. et al. Fructose consumption as a risk factor for non-alcoholic fatty liver disease. J. Hepatol. 48, 993–999 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Vos, M. B. & Lavine, J. E. Dietary fructose in nonalcoholic fatty liver disease. Hepatology 57, 2525–2531 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Basaranoglu, M., Basaranoglu, G., Sabuncu, T. & Senturk, H. Fructose as a key player in the development of fatty liver disease. World J. Gastroenterol. 19, 1166–1172 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Casals-Casas, C. & Desvergne, B. Endocrine disruptors: from endocrine to metabolic disruption. Annu. Rev. Physiol. 73, 135–162 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Heindel, J. J., Newbold, R. & Schug, T. T. Endocrine disruptors and obesity. Nat. Rev. Endocrinol. 11, 653–661 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Grun, F. & Blumberg, B. Perturbed nuclear receptor signaling by environmental obesogens as emerging factors in the obesity crisis. Rev. Endocr. Metab. Disord. 8, 161–171 (2007).

    Article  PubMed  Google Scholar 

  35. Alonso-Magdalena, P., Quesada, I. & Nadal, A. Endocrine disruptors in the etiology of type 2 diabetes mellitus. Nat. Rev. Endocrinol. 7, 346–353 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Vom Saal, F. S., Nagel, S. C., Coe, B. L., Angle, B. M. & Taylor, J. A. The estrogenic endocrine disrupting chemical bisphenol A (BPA) and obesity. Mol. Cell. Endocrinol. 354, 74–84 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lonard, D. M. & O'Malley, B. W. Nuclear receptor coregulators: judges, juries, and executioners of cellular regulation. Mol. Cell 27, 691–700 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Foulds, C. E. et al. Proteomic analysis of coregulators bound to ERα on DNA and nucleosomes reveals coregulator dynamics. Mol. Cell 51, 185–199 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Smith, C. L. & O'Malley, B. W. Coregulator function: a key to understanding tissue specificity of selective receptor modulators. Endocr. Rev. 25, 45–71 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Routledge, E. J., White, R., Parker, M. G. & Sumpter, J. P. Differential effects of xenoestrogens on coactivator recruitment by estrogen receptor (ER) α and ERβ. J. Biol. Chem. 275, 35986–35993 (2000).

    Article  CAS  PubMed  Google Scholar 

  41. Levin, E. R. Cell localization, physiology, and nongenomic actions of estrogen receptors. J. Appl. Physiol. 91, 1860–1867 (2001).

    Article  CAS  PubMed  Google Scholar 

  42. Trevino, L. S. & Weigel, N. L. Phosphorylation: a fundamental regulator of steroid receptor action. Trends Endocrinol. Metab. 24, 515–524 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Hammes, S. R. & Levin, E. R. Minireview: recent advances in extranuclear steroid receptor actions. Endocrinology 152, 4489–4495 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Hammes, S. R. & Davis, P. J. Overlapping nongenomic and genomic actions of thyroid hormone and steroids. Best Pract. Res. Clin. Endocrinol. Metab. 29, 581–593 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Endo, Y. et al. Thiazolidinediones enhance sodium-coupled bicarbonate absorption from renal proximal tubules via PPARγ-dependent nongenomic signaling. Cell Metab. 13, 550–561 (2011).

    Article  CAS  PubMed  Google Scholar 

  46. Zhang, X. K. et al. Regulation of the nongenomic actions of retinoid X receptor-α by targeting the coregulator-binding sites. Acta Pharmacol. Sin. 36, 102–112 (2015).

    Article  PubMed  CAS  Google Scholar 

  47. Davis, P. J., Goglia, F. & Leonard, J. L. Nongenomic actions of thyroid hormone. Nat. Rev. Endocrinol. 12, 111–121 (2016).

    Article  CAS  PubMed  Google Scholar 

  48. Al Tanoury, Z., Piskunov, A. & Rochette-Egly, C. Vitamin A and retinoid signaling: genomic and nongenomic effects. J. Lipid Res. 54, 1761–1775 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bookout, A. L. et al. Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network. Cell 126, 789–799 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Liu, Q. et al. In-depth proteomic characterization of endogenous nuclear receptors in mouse liver. Mol. Cell. Proteomics 12, 473–484 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Cave, M. C. et al. Nuclear receptors and nonalcoholic fatty liver disease. Biochim. Biophys. Acta 1859, 1083–1099 (2016). This review highlights the key NRs implicated in NAFLD progression.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ballestri, S., Nascimbeni, F., Romagnoli, D., Baldelli, E. & Lonardo, A. The role of nuclear receptors in the pathophysiology, natural course, and drug treatment of NAFLD in humans. Adv. Ther. 33, 291–319 (2016).

    Article  CAS  PubMed  Google Scholar 

  53. Gross, B., Pawlak, M., Lefebvre, P. & Staels, B. PPARs in obesity-induced T2DM, dyslipidaemia and NAFLD. Nat. Rev. Endocrinol. 13, 36–49 (2017).

    Article  CAS  PubMed  Google Scholar 

  54. Wahli, W. & Michalik, L. PPARs at the crossroads of lipid signaling and inflammation. Trends Endocrinol. Metab. 23, 351–363 (2012).

    Article  CAS  PubMed  Google Scholar 

  55. Timsit, Y. E. & Negishi, M. CAR and PXR: the xenobiotic-sensing receptors. Steroids 72, 231–246 (2007).

    Article  CAS  PubMed  Google Scholar 

  56. Chai, S. C., Cherian, M. T., Wang, Y. M. & Chen, T. Small-molecule modulators of PXR and CAR. Biochim. Biophys. Acta 1859, 1141–1154 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Moore, L. B. et al. Orphan nuclear receptors constitutive androstane receptor and pregnane X receptor share xenobiotic and steroid ligands. J. Biol. Chem. 275, 15122–15127 (2000).

    Article  CAS  PubMed  Google Scholar 

  58. Tzameli, I., Pissios, P., Schuetz, E. G. & Moore, D. D. The xenobiotic compound 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene is an agonist ligand for the nuclear receptor CAR. Mol. Cell. Biol. 20, 2951–2958 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Maglich, J. M. et al. Identification of a novel human constitutive androstane receptor (CAR) agonist and its use in the identification of CAR target genes. J. Biol. Chem. 278, 17277–17283 (2003).

    Article  CAS  PubMed  Google Scholar 

  60. Jones, S. A. et al. The pregnane X receptor: a promiscuous xenobiotic receptor that has diverged during evolution. Mol. Endocrinol. 14, 27–39 (2000).

    Article  CAS  PubMed  Google Scholar 

  61. Lehmann, J. M. et al. The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. J. Clin. Invest. 102, 1016–1023 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Blumberg, B. et al. SXR, a novel steroid and xenobiotic-sensing nuclear receptor. Genes Dev. 12, 3195–3205 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Bertilsson, G. et al. Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction. Proc. Natl Acad. Sci. USA 95, 12208–12213 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Dong, B. et al. Activation of nuclear receptor CAR ameliorates diabetes and fatty liver disease. Proc. Natl Acad. Sci. USA 106, 18831–18836 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhou, J. et al. A novel pregnane X receptor-mediated and sterol regulatory element-binding protein-independent lipogenic pathway. J. Biol. Chem. 281, 15013–15020 (2006).

    Article  CAS  PubMed  Google Scholar 

  66. Schultz, J. R. et al. Role of LXRs in control of lipogenesis. Genes Dev. 14, 2831–2838 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Watanabe, M. et al. Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1c. J. Clin. Invest. 113, 1408–1418 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Perra, A. et al. Thyroid hormone (T3) and TRβ agonist GC-1 inhibit/reverse nonalcoholic fatty liver in rats. FASEB J. 22, 2981–2989 (2008).

    Article  CAS  PubMed  Google Scholar 

  69. Martagon, A. J., Lin, J. Z., Cimini, S. L., Webb, P. & Phillips, K. J. The amelioration of hepatic steatosis by thyroid hormone receptor agonists is insufficient to restore insulin sensitivity in ob/ob mice. PLoS ONE 10, e0122987 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Lopez-Velazquez, J. A., Carrillo-Cordova, L. D., Chavez-Tapia, N. C., Uribe, M. & Mendez-Sanchez, N. Nuclear receptors in nonalcoholic fatty liver disease. J. Lipids 2012, 139875 (2012).

    Article  PubMed  CAS  Google Scholar 

  71. Sun, M., Cui, W., Woody, S. K. & Staudinger, J. L. Pregnane X receptor modulates the inflammatory response in primary cultures of hepatocytes. Drug Metab. Dispos. 43, 335–343 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Mouchiroud, L., Eichner, L. J., Shaw, R. J. & Auwerx, J. Transcriptional coregulators: fine-tuning metabolism. Cell Metab. 20, 26–40 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. York, B. et al. Research resource: tissue- and pathway-specific metabolomic profiles of the steroid receptor coactivator (SRC) family. Mol. Endocrinol. 27, 366–380 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Chopra, A. R. et al. Absence of the SRC-2 coactivator results in a glycogenopathy resembling Von Gierke's disease. Science 322, 1395–1399 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Ma, X. et al. Deletion of steroid receptor coactivator-3 gene ameliorates hepatic steatosis. J. Hepatol. 55, 445–452 (2011).

    Article  CAS  PubMed  Google Scholar 

  76. Stashi, E., York, B. & O'Malley, B. W. Steroid receptor coactivators: servants and masters for control of systems metabolism. Trends Endocrinol. Metab. 25, 337–347 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Rollins, D. A., Coppo, M. & Rogatsky, I. Minireview: nuclear receptor coregulators of the p160 family: insights into inflammation and metabolism. Mol. Endocrinol. 29, 502–517 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Leone, T. C. et al. PGC-1α deficiency causes multi-system energy metabolic derangements: muscle dysfunction, abnormal weight control and hepatic steatosis. PLoS Biol. 3, e101 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Sonoda, J., Mehl, I. R., Chong, L. W., Nofsinger, R. R. & Evans, R. M. PGC-1β controls mitochondrial metabolism to modulate circadian activity, adaptive thermogenesis, and hepatic steatosis. Proc. Natl Acad. Sci. USA 104, 5223–5228 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Lelliott, C. J. et al. Ablation of PGC-1β results in defective mitochondrial activity, thermogenesis, hepatic function, and cardiac performance. PLoS Biol. 4, e369 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Li, S. et al. Genome-wide coactivation analysis of PGC-1α identifies BAF60a as a regulator of hepatic lipid metabolism. Cell Metab. 8, 105–117 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Yuan, C. X., Ito, M., Fondell, J. D., Fu, Z. Y. & Roeder, R. G. The TRAP220 component of a thyroid hormone receptor-associated protein (TRAP) coactivator complex interacts directly with nuclear receptors in a ligand-dependent fashion. Proc. Natl Acad. Sci. USA 95, 7939–7944 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Bai, L. et al. Transcription coactivator mediator subunit MED1 is required for the development of fatty liver in the mouse. Hepatology 53, 1164–1174 (2011).

    Article  CAS  PubMed  Google Scholar 

  84. Guenther, M. G. et al. A core SMRT corepressor complex containing HDAC3 and TBL1, a WD40-repeat protein linked to deafness. Genes Dev. 14, 1048–1057 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Yoon, H. G. et al. Purification and functional characterization of the human N-CoR complex: the roles of HDAC3, TBL1 and TBLR1. EMBO J. 22, 1336–1346 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Zhang, J., Kalkum, M., Chait, B. T. & Roeder, R. G. The N-CoR–HDAC3 nuclear receptor corepressor complex inhibits the JNK pathway through the integral subunit GPS2. Mol. Cell 9, 611–623 (2002).

    Article  CAS  PubMed  Google Scholar 

  87. Sun, Z. et al. Hepatic Hdac3 promotes gluconeogenesis by repressing lipid synthesis and sequestration. Nat. Med. 18, 934–942 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Sun, Z. et al. Deacetylase-independent function of HDAC3 in transcription and metabolism requires nuclear receptor corepressor. Mol. Cell 52, 769–782 (2013).

    Article  CAS  PubMed  Google Scholar 

  89. Leonardsson, G. et al. Nuclear receptor corepressor RIP140 regulates fat accumulation. Proc. Natl Acad. Sci. USA 101, 8437–8442 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Berriel Diaz, M. et al. Nuclear receptor cofactor receptor interacting protein 140 controls hepatic triglyceride metabolism during wasting in mice. Hepatology 48, 782–791 (2008).

    Article  PubMed  CAS  Google Scholar 

  91. Song, Y. et al. Ligand-dependent corepressor acts as a novel corepressor of thyroid hormone receptor and represses hepatic lipogenesis in mice. J. Hepatol. 56, 248–254 (2012).

    Article  CAS  PubMed  Google Scholar 

  92. Lee, J. M. et al. Ursodeoxycholic acid inhibits liver X receptor α-mediated hepatic lipogenesis via induction of the nuclear corepressor SMILE. J. Biol. Chem. 289, 1079–1091 (2014).

    Article  CAS  PubMed  Google Scholar 

  93. Zheng, F. F., Wu, R. C., Smith, C. L. & O'Malley, B. W. Rapid estrogen-induced phosphorylation of the SRC-3 coactivator occurs in an extranuclear complex containing estrogen receptor. Mol. Cell. Biol. 25, 8273–8284 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wu, R. C. et al. Selective phosphorylations of the SRC-3/AIB1 coactivator integrate genomic reponses to multiple cellular signaling pathways. Mol. Cell 15, 937–949 (2004).

    Article  CAS  PubMed  Google Scholar 

  95. York, B. et al. Reprogramming the posttranslational code of SRC-3 confers a switch in mammalian systems biology. Proc. Natl Acad. Sci. USA 107, 11122–11127 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Jo, Y. S. et al. Phosphorylation of the nuclear receptor corepressor 1 by protein kinase B switches its corepressor targets in the liver in mice. Hepatology 62, 1606–1618 (2015).

    Article  CAS  PubMed  Google Scholar 

  97. le Maire, A., Bourguet, W. & Balaguer, P. A structural view of nuclear hormone receptor: endocrine disruptor interactions. Cell. Mol. Life Sci. 67, 1219–1237 (2010).

    Article  PubMed  CAS  Google Scholar 

  98. Swedenborg, E., Ruegg, J., Makela, S. & Pongratz, I. Endocrine disruptive chemicals: mechanisms of action and involvement in metabolic disorders. J. Mol. Endocrinol. 43, 1–10 (2009).

    Article  CAS  PubMed  Google Scholar 

  99. Rich, R. L. et al. Kinetic analysis of estrogen receptor/ligand interactions. Proc. Natl Acad. Sci. USA 99, 8562–8567 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Grun, F. et al. Endocrine-disrupting organotin compounds are potent inducers of adipogenesis in vertebrates. Mol. Endocrinol. 20, 2141–2155 (2006). This paper forms the basis for the obesogen hypothesis, as it shows that the EDC TBT promotes adipogenesis by activating PPARγ and RXRα.

    Article  CAS  PubMed  Google Scholar 

  101. Mellor, C. L., Steinmetz, F. P. & Cronin, M. T. The identification of nuclear receptors associated with hepatic steatosis to develop and extend adverse outcome pathways. Crit. Rev. Toxicol. 46, 138–152 (2016).

    Article  CAS  PubMed  Google Scholar 

  102. Wahlang, B. et al. Polychlorinated biphenyl-xenobiotic nuclear receptor interactions regulate energy metabolism, behavior, and inflammation in non-alcoholic-steatohepatitis. Toxicol. Sci. 149, 396–410 (2016).

    Article  CAS  PubMed  Google Scholar 

  103. Al-Eryani, L. et al. Identification of environmental chemicals associated with the development of toxicant-associated fatty liver disease in rodents. Toxicol. Pathol. 43, 482–497 (2015).

    Article  CAS  PubMed  Google Scholar 

  104. Tomaszewski, K. E., Montgomery, C. A. & Melnick, R. L. Modulation of 2,3,7,8-tetrachlorodibenzo-p-dioxin toxicity in F344 rats by di(2-ethylhexyl)phthalate. Chem. Biol. Interact. 65, 205–222 (1988).

    Article  CAS  PubMed  Google Scholar 

  105. Shan, Q., Huang, F., Wang, J. & Du, Y. Effects of co-exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin and polychlorinated biphenyls on nonalcoholic fatty liver disease in mice. Environ. Toxicol. 30, 1364–1374 (2015). This paper reports how exposure to two different types of EDC can have a synergistic effect in producing a NAFLD phenotype in mice.

    Article  CAS  PubMed  Google Scholar 

  106. Wei, J. et al. Perinatal exposure to bisphenol A exacerbates nonalcoholic steatohepatitis-like phenotype in male rat offspring fed on a high-fat diet. J. Endocrinol. 222, 313–325 (2014). This is the first of two seminal studies reporting that perinatal exposure to BPA combined with a HFD after weaning leads to more severe hepatic steatosis and increases inflammation in male, but not female, offspring (see also reference 107).

    Article  CAS  PubMed  Google Scholar 

  107. Strakovsky, R. S. et al. Developmental bisphenol A (BPA) exposure leads to sex-specific modification of hepatic gene expression and epigenome at birth that may exacerbate high-fat diet-induced hepatic steatosis. Toxicol. Appl. Pharmacol. 284, 101–112 (2015). This is the second seminal BPA study to report that perinatal exposure to BPA augments HFD-induced hepatic steatosis; the authors show that exposure to BPA alters the DNA methylation and histone modifications of a gene that encodes an enzyme that is important in the β-oxidation pathway (see also reference 106).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Tan, X. et al. High fat diet feeding exaggerates perfluorooctanoic acid-induced liver injury in mice via modulating multiple metabolic pathways. PLoS ONE 8, e61409 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Jin, Y. et al. Chronic exposure of mice to environmental endocrine-disrupting chemicals disturbs their energy metabolism. Toxicol. Lett. 225, 392–400 (2014).

    Article  CAS  PubMed  Google Scholar 

  110. Wahlang, B. et al. Polychlorinated biphenyl 153 is a diet-dependent obesogen that worsens nonalcoholic fatty liver disease in male C57BL6/J mice. J. Nutr. Biochem. 24, 1587–1595 (2013). This paper reports that the exposure of rodents to some EDCs (such as PCB153) does not lead to NAFLD when they are administered alone but can promote that induced by a HFD.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Wahlang, B. et al. Evaluation of Aroclor 1260 exposure in a mouse model of diet-induced obesity and non-alcoholic fatty liver disease. Toxicol. Appl. Pharmacol. 279, 380–390 (2014). This paper reports that the exposure of rodents to some EDCs (such as Aroclor 1260) might not worsen HFD-induced hepatic steatosis but instead induce a NASH-like phenotype.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Pazderova-Vejlupkova, J., Lukas, E., Nemcova, M., Pickova, J. & Jirasek, L. The development and prognosis of chronic intoxication by tetrachlordibenzo-p-dioxin in men. Arch. Environ. Health 36, 5–11 (1981).

    Article  CAS  PubMed  Google Scholar 

  113. Obika, M. & Noguchi, H. Diagnosis and evaluation of nonalcoholic fatty liver disease. Exp. Diabetes Res. 2012, 145754 (2012).

    Article  PubMed  CAS  Google Scholar 

  114. Waddington, C. H. Canalization of development and the inheritance of acquired characters. Nature 150, 563–565 (1942).

    Article  Google Scholar 

  115. Holliday, R. & Pugh, J. E. DNA modification mechanisms and gene activity during development. Science 187, 226–232 (1975).

    Article  CAS  PubMed  Google Scholar 

  116. Riggs, A. D. X inactivation, differentiation, and DNA methylation. Cytogenet. Cell Genet. 14, 9–25 (1975).

    Article  CAS  PubMed  Google Scholar 

  117. Bell, A. C. & Felsenfeld, G. Methylation of a CTCF-dependent boundary controls imprinted expression of the Igf2 gene. Nature 405, 482–485 (2000).

    Article  CAS  PubMed  Google Scholar 

  118. Bell, A. C., West, A. G. & Felsenfeld, G. The protein CTCF is required for the enhancer blocking activity of vertebrate insulators. Cell 98, 387–396 (1999).

    Article  CAS  PubMed  Google Scholar 

  119. Watt, F. & Molloy, P. L. Cytosine methylation prevents binding to DNA of a HeLa cell transcription factor required for optimal expression of the adenovirus major late promoter. Genes Dev. 2, 1136–1143 (1988).

    Article  CAS  PubMed  Google Scholar 

  120. Fournier, A., Sasai, N., Nakao, M. & Defossez, P. A. The role of methyl-binding proteins in chromatin organization and epigenome maintenance. Brief. Funct. Genom. 11, 251–264 (2012).

    Article  CAS  Google Scholar 

  121. Luger, K., Mader, A. W., Richmond, R. K., Sargent, D. F. & Richmond, T. J. Crystal structure of the nucleosome core particle at 2.8 Å resolution. Nature 389, 251–260 (1997).

    Article  CAS  PubMed  Google Scholar 

  122. Lee, J. S., Smith, E. & Shilatifard, A. The language of histone crosstalk. Cell 142, 682–685 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Jenuwein, T. & Allis, C. D. Translating the histone code. Science 293, 1074–1080 (2001).

    Article  CAS  PubMed  Google Scholar 

  124. Hong, L., Schroth, G. P., Matthews, H. R., Yau, P. & Bradbury, E. M. Studies of the DNA binding properties of histone H4 amino terminus. Thermal denaturation studies reveal that acetylation markedly reduces the binding constant of the H4 “tail” to DNA. J. Biol. Chem. 268, 305–314 (1993).

    Article  CAS  PubMed  Google Scholar 

  125. Yang, X. J. & Seto, E. Collaborative spirit of histone deacetylases in regulating chromatin structure and gene expression. Curr. Opin. Genet. Dev. 13, 143–153 (2003).

    Article  CAS  PubMed  Google Scholar 

  126. Yang, X. J. & Seto, E. HATs and HDACs: from structure, function and regulation to novel strategies for therapy and prevention. Oncogene 26, 5310–5318 (2007).

    Article  CAS  PubMed  Google Scholar 

  127. Noureddin, M., Mato, J. M. & Lu, S. C. Nonalcoholic fatty liver disease: update on pathogenesis, diagnosis, treatment and the role of S-adenosylmethionine. Exp. Biol. Med. (Maywood) 240, 809–820 (2015).

    Article  CAS  Google Scholar 

  128. Lu, S. C. & Mato, J. M. S-Adenosylmethionine in liver health, injury, and cancer. Physiol. Rev. 92, 1515–1542 (2012).

    Article  CAS  PubMed  Google Scholar 

  129. Lu, S. C. et al. Methionine adenosyltransferase 1A knockout mice are predisposed to liver injury and exhibit increased expression of genes involved in proliferation. Proc. Natl Acad. Sci. USA 98, 5560–5565 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Martinez-Chantar, M. L. et al. Loss of the glycine N-methyltransferase gene leads to steatosis and hepatocellular carcinoma in mice. Hepatology 47, 1191–1199 (2008).

    Article  CAS  PubMed  Google Scholar 

  131. Varela-Rey, M. et al. Fatty liver and fibrosis in glycine N-methyltransferase knockout mice is prevented by nicotinamide. Hepatology 52, 105–114 (2010).

    Article  CAS  PubMed  Google Scholar 

  132. Augoustides-Savvopoulou, P. et al. Glycine N-methyltransferase deficiency: a new patient with a novel mutation. J. Inherit. Metab. Dis. 26, 745–759 (2003).

    Article  CAS  PubMed  Google Scholar 

  133. Mudd, S. H. et al. Glycine N-methyltransferase deficiency: a novel inborn error causing persistent isolated hypermethioninaemia. J. Inherit. Metab. Dis. 24, 448–464 (2001).

    Article  CAS  PubMed  Google Scholar 

  134. Pogribny, I. P. et al. Hepatic epigenetic phenotype predetermines individual susceptibility to hepatic steatosis in mice fed a lipogenic methyl-deficient diet. J. Hepatol. 51, 176–186 (2009). This paper provides data that mechanistically link epigenetic alterations (namely, DNA methylation and certain histone modifications) to the pathogenesis of hepatic steatosis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Tryndyak, V. et al. Interstrain differences in the severity of liver injury induced by a choline- and folate-deficient diet in mice are associated with dysregulation of genes involved in lipid metabolism. FASEB J. 26, 4592–4602 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Tryndyak, V. P. et al. Status of hepatic DNA methylome predetermines and modulates the severity of non-alcoholic fatty liver injury in mice. BMC Genomics 17, 298 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  137. Tryndyak, V. P. et al. Coupling global methylation and gene expression profiles reveal key pathophysiological events in liver injury induced by a methyl-deficient diet. Mol. Nutr. Food Res. 55, 411–418 (2011).

    Article  CAS  PubMed  Google Scholar 

  138. Lee, J. H., Friso, S. & Choi, S. W. Epigenetic mechanisms underlying the link between non-alcoholic fatty liver diseases and nutrition. Nutrients 6, 3303–3325 (2014). This review highlights the important concept that nutrient 'signals' can have epigenomic effects (on DNA methylation and histone modifications) that might contribute to NAFLD progression.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Purushotham, A. et al. Hepatocyte-specific deletion of SIRT1 alters fatty acid metabolism and results in hepatic steatosis and inflammation. Cell Metab. 9, 327–338 (2009). This paper reports that the loss of a specific HDAC (SIRT1) in the liver results in an NAFLD phenotype through the inactivation of the co-activator PGC1α.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Feng, D. et al. A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism. Science 331, 1315–1319 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Mikula, M., Majewska, A., Ledwon, J. K., Dzwonek, A. & Ostrowski, J. Obesity increases histone H3 lysine 9 and 18 acetylation at Tnfa and Ccl2 genes in mouse liver. Int. J. Mol. Med. 34, 1647–1654 (2014).

    Article  CAS  PubMed  Google Scholar 

  142. Aagaard-Tillery, K. M. et al. Developmental origins of disease and determinants of chromatin structure: maternal diet modifies the primate fetal epigenome. J. Mol. Endocrinol. 41, 91–102 (2008). This paper reports that a maternal HFD leads to increased liver levels of triglycerides and epigenomic alternations (specifically histone H3 lysine 14 acetylation) in Japanese macaque fetuses.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Suter, M. A. et al. A maternal high-fat diet modulates fetal SIRT1 histone and protein deacetylase activity in nonhuman primates. FASEB J. 26, 5106–5114 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Jirtle, R. L. The Agouti mouse: a biosensor for environmental epigenomics studies investigating the developmental origins of health and disease. Epigenomics 6, 447–450 (2014).

    Article  CAS  PubMed  Google Scholar 

  145. Dolinoy, D. C., Das, R., Weidman, J. R. & Jirtle, R. L. Metastable epialleles, imprinting, and the fetal origins of adult diseases. Pediatr. Res. 61, 30R–37R (2007).

    Article  PubMed  Google Scholar 

  146. Miltenberger, R. J., Mynatt, R. L., Wilkinson, J. E. & Woychik, R. P. The role of the agouti gene in the yellow obese syndrome. J. Nutr. 127, 1902S–1907S (1997).

    Article  CAS  PubMed  Google Scholar 

  147. Morgan, H. D., Sutherland, H. G., Martin, D. I. & Whitelaw, E. Epigenetic inheritance at the agouti locus in the mouse. Nat. Genet. 23, 314–318 (1999).

    Article  CAS  PubMed  Google Scholar 

  148. Dolinoy, D. C., Weidman, J. R., Waterland, R. A. & Jirtle, R. L. Maternal genistein alters coat color and protects Avy mouse offspring from obesity by modifying the fetal epigenome. Environ. Health Perspect. 114, 567–572 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Dolinoy, D. C., Huang, D. & Jirtle, R. L. Maternal nutrient supplementation counteracts bisphenol A-induced DNA hypomethylation in early development. Proc. Natl Acad. Sci. USA 104, 13056–13061 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Murphy, S. K. et al. Relationship between methylome and transcriptome in patients with nonalcoholic fatty liver disease. Gastroenterology 145, 1076–1087 (2013).

    Article  CAS  PubMed  Google Scholar 

  151. Zeybel, M. et al. Differential DNA methylation of genes involved in fibrosis progression in non-alcoholic fatty liver disease and alcoholic liver disease. Clin. Epigenetics 7, 25 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. Barker, D. J. Sir Richard Doll Lecture. Developmental origins of chronic disease. Public Health 126, 185–189 (2012).

    Article  CAS  PubMed  Google Scholar 

  153. Bruce, K. D. et al. Maternal high-fat feeding primes steatohepatitis in adult mice offspring, involving mitochondrial dysfunction and altered lipogenesis gene expression. Hepatology 50, 1796–1808 (2009). This paper provides a link between maternal HFD and NAFLD progression to steatohepatitis in adult rodent offspring.

    Article  CAS  PubMed  Google Scholar 

  154. Elahi, M. M. et al. Long-term maternal high-fat feeding from weaning through pregnancy and lactation predisposes offspring to hypertension, raised plasma lipids and fatty liver in mice. Br. J. Nutr. 102, 514–519 (2009).

    Article  CAS  PubMed  Google Scholar 

  155. Gregorio, B. M., Souza-Mello, V., Carvalho, J. J., Mandarim-de-Lacerda, C. A. & Aguila, M. B. Maternal high-fat intake predisposes nonalcoholic fatty liver disease in C57BL/6 offspring. Am. J. Obstet. Gynecol. 203, 495.e1–495.e8 (2010).

    Article  CAS  Google Scholar 

  156. Oben, J. A. et al. Maternal obesity during pregnancy and lactation programs the development of offspring non-alcoholic fatty liver disease in mice. J. Hepatol. 52, 913–920 (2010).

    Article  CAS  PubMed  Google Scholar 

  157. Ashino, N. G. et al. Maternal high-fat feeding through pregnancy and lactation predisposes mouse offspring to molecular insulin resistance and fatty liver. J. Nutr. Biochem. 23, 341–348 (2012).

    Article  CAS  PubMed  Google Scholar 

  158. Li, J. et al. Accumulation of endoplasmic reticulum stress and lipogenesis in the liver through generational effects of high fat diets. J. Hepatol. 56, 900–907 (2012).

    Article  CAS  PubMed  Google Scholar 

  159. Mouralidarane, A. et al. Maternal obesity programs offspring nonalcoholic fatty liver disease by innate immune dysfunction in mice. Hepatology 58, 128–138 (2013).

    Article  CAS  PubMed  Google Scholar 

  160. Kjaergaard, M., Nilsson, C., Rosendal, A., Nielsen, M. O. & Raun, K. Maternal chocolate and sucrose soft drink intake induces hepatic steatosis in rat offspring associated with altered lipid gene expression profile. Acta Physiol. (Oxf.) 210, 142–153 (2014).

    Article  CAS  Google Scholar 

  161. Pruis, M. G. et al. Maternal western diet primes non-alcoholic fatty liver disease in adult mouse offspring. Acta Physiol. (Oxf.) 210, 215–227 (2014).

    Article  CAS  Google Scholar 

  162. Chen, J. P. et al. Fetal and infant exposure to the Chinese famine increases the risk of fatty liver disease in Chongqing, China. J. Gastroenterol. Hepatol. 31, 200–205 (2016).

    Article  PubMed  Google Scholar 

  163. Waterland, R. A. & Jirtle, R. L. Transposable elements: targets for early nutritional effects on epigenetic gene regulation. Mol. Cell. Biol. 23, 5293–5300 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Kim, J. M., Hong, K., Lee, J. H., Lee, S. & Chang, N. Effect of folate deficiency on placental DNA methylation in hyperhomocysteinemic rats. J. Nutr. Biochem. 20, 172–176 (2009).

    Article  CAS  PubMed  Google Scholar 

  165. Lillycrop, K. A., Phillips, E. S., Jackson, A. A., Hanson, M. A. & Burdge, G. C. Dietary protein restriction of pregnant rats induces and folic acid supplementation prevents epigenetic modification of hepatic gene expression in the offspring. J. Nutr. 135, 1382–1386 (2005).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge K. Phillips, M. Feingold and D. Moore for helpful discussions on nonalcoholic fatty liver disease, and the National Institute of Environmental Health Sciences for support (grants 1R01ES023206, P30ES023512 and U01ES026719 to C.L.W.).

Author information

Authors and Affiliations

Authors

Contributions

C.E.F., L.S.T., B.Y. and C.L.W. researched data for the article, made substantial contributions to discussions about the content, wrote the article, and reviewed and/or edited the manuscript before submission.

Corresponding author

Correspondence to Cheryl L. Walker.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information S1 (table)

EDCs reported to induce a NAFLD phenotype in rodents. (PDF 876 kb)

Supplementary information S2 (table)

EDCs associated with liver dysfunction and potential NAFLD in humans. (PDF 396 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Foulds, C., Treviño, L., York, B. et al. Endocrine-disrupting chemicals and fatty liver disease. Nat Rev Endocrinol 13, 445–457 (2017). https://doi.org/10.1038/nrendo.2017.42

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrendo.2017.42

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing