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.

  • Article
  • Published:

Multigenerational epigenetic adaptation of the hepatic wound-healing response

A Corrigendum to this article was published on 05 October 2012

This article has been updated

Abstract

We investigated whether ancestral liver damage leads to heritable reprogramming of hepatic wound healing in male rats. We found that a history of liver damage corresponds with transmission of an epigenetic suppressive adaptation of the fibrogenic component of wound healing to the male F1 and F2 generations. Underlying this adaptation was less generation of liver myofibroblasts, higher hepatic expression of the antifibrogenic factor peroxisome proliferator-activated receptor γ (PPAR-γ) and lower expression of the profibrogenic factor transforming growth factor β1 (TGF-β1) compared to rats without this adaptation. Remodeling of DNA methylation and histone acetylation underpinned these alterations in gene expression. Sperm from rats with liver fibrosis were enriched for the histone variant H2A.Z and trimethylation of histone H3 at Lys27 (H3K27me3) at PPAR-γ chromatin. These modifications to the sperm chromatin were transmittable by adaptive serum transfer from fibrotic rats to naive rats and similar modifications were induced in mesenchymal stem cells exposed to conditioned media from cultured rat or human myofibroblasts. Thus, it is probable that a myofibroblast-secreted soluble factor stimulates heritable epigenetic signatures in sperm so that the resulting offspring better adapt to future fibrogenic hepatic insults. Adding possible relevance to humans, we found that people with mild liver fibrosis have hypomethylation of the PPARG promoter compared to others with severe fibrosis.

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 presence of liver injury in male ancestors reduces liver fibrogenesis in F2-generation male offspring.
Figure 2: Paternally transmitted adaptation of the fibrogenic response in chronic injury is mediated by a low number of α-SMA–positive myofibroblasts in the liver.
Figure 3: Protected rats have altered expression of profibrogenic and antifibrogenic genes in their livers.
Figure 4: The fibrogenic response that is influenced by ancestral injury is limited to the liver.
Figure 5: Altered expression of profibrogenic and antifibrogenic genes in the livers of protected rats is underpinned by differences in DNA methylation.
Figure 6: Extrahepatic transmission of epigenetic modifications and evidence for modifications in DNA methylation at fibrogenic regulator genes that are associated with the progression of liver disease in humans.

Similar content being viewed by others

Change history

  • 05 October 2012

     In the version of this article initially published, in Figure 6f the left-hand graph was an inadvertent duplication of the right-hand graph. The error does not alter the overall conclusions of the paper. The figure has been corrected in the HTML and PDF versions of the article.

References

  1. Burd, D.A. et al. Fetal wound healing: an in vitro explant model. J. Pediatr. Surg. 25, 898–901 (1990).

    Article  CAS  Google Scholar 

  2. Harrison, M.R. & Adzick, N.S. The fetus as a patient. Surgical considerations. Ann. Surg. 213, 279–291, discussion 277–278 (1991).

    Article  CAS  Google Scholar 

  3. Kalluri, R. & Zeisberg, M. Fibroblasts in cancer. Nat. Rev. Cancer 6, 392–401 (2006).

    Article  CAS  Google Scholar 

  4. King, T.E. Jr., Pardo, A. & Selman, M. Idiopathic pulmonary fibrosis. Lancet 378, 1949–1961 (2011).

    Article  Google Scholar 

  5. Pinzani, M., Rombouts, K. & Colagrande, S. Fibrosis in chronic liver diseases: diagnosis and management. J. Hepatol. 42 (suppl.), S22–S36 (2005).

    Article  Google Scholar 

  6. Matteoni, C.A. et al. Nonalcoholic fatty liver disease: a spectrum of clinical and pathological severity. Gastroenterology 116, 1413–1419 (1999).

    Article  CAS  Google Scholar 

  7. Poynard, T., Bedossa, P. & Opolon, P. Natural history of liver fibrosis progression in patients with chronic hepatitis C. The OBSVIRC, METAVIR, CLINIVIR, and DOSVIRC groups. Lancet 349, 825–832 (1997).

    Article  CAS  Google Scholar 

  8. Teli, M.R., Day, C.P., Burt, A.D., Bennett, M.K. & James, O.F. Determinants of progression to cirrhosis or fibrosis in pure alcoholic fatty liver. Lancet 346, 987–990 (1995).

    Article  CAS  Google Scholar 

  9. Fischle, W., Wang, Y. & Allis, C.D. Histone and chromatin cross-talk. Curr. Opin. Cell Biol. 15, 172–183 (2003).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  11. Lavrov, S.A. & Kibanov, M.V. Noncoding RNAs and chromatin structure. Biochemistry (Mosc.) 72, 1422–1438 (2007).

    Article  CAS  Google Scholar 

  12. Strahl, B.D. & Allis, C.D. The language of covalent histone modifications. Nature 403, 41–45 (2000).

    Article  CAS  Google Scholar 

  13. Umlauf, D., Fraser, P. & Nagano, T. The role of long non-coding RNAs in chromatin structure and gene regulation: variations on a theme. Biol. Chem. 389, 323–331 (2008).

    Article  CAS  Google Scholar 

  14. Barker, D.J. The origins of the developmental origins theory. J. Intern. Med. 261, 412–417 (2007).

    Article  CAS  Google Scholar 

  15. Recknagel, R.O., Glende, E.A. Jr., Dolak, J.A. & Waller, R.L. Mechanisms of carbon tetrachloride toxicity. Pharmacol. Ther. 43, 139–154 (1989).

    Article  CAS  Google Scholar 

  16. Williams, A.T. & Burk, R.F. Carbon tetrachloride hepatotoxicity: an example of free radical–mediated injury. Semin. Liver Dis. 10, 279–284 (1990).

    Article  CAS  Google Scholar 

  17. Raucy, J.L., Kraner, J.C. & Lasker, J.M. Bioactivation of halogenated hydrocarbons by cytochrome P4502E1. Crit. Rev. Toxicol. 23, 1–20 (1993).

    Article  CAS  Google Scholar 

  18. Friedman, S.L. Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol. Rev. 88, 125–172 (2008).

    Article  CAS  Google Scholar 

  19. Friedman, S.L. Mechanisms of hepatic fibrogenesis. Gastroenterology 134, 1655–1669 (2008).

    Article  CAS  Google Scholar 

  20. Yokoi, Y. et al. Immunocytochemical detection of desmin in fat-storing cells (Ito cells). Hepatology 4, 709–714 (1984).

    Article  CAS  Google Scholar 

  21. Hazra, S. et al. Peroxisome proliferator-activated receptor gamma induces a phenotypic switch from activated to quiescent hepatic stellate cells. J. Biol. Chem. 279, 11392–11401 (2004).

    Article  CAS  Google Scholar 

  22. Mann, J. et al. MeCP2 controls an epigenetic pathway that promotes myofibroblast transdifferentiation and fibrosis. Gastroenterology 138, 705–714 (2010).

    Article  CAS  Google Scholar 

  23. Miyahara, T. et al. Peroxisome proliferator-activated receptors and hepatic stellate cell activation. J. Biol. Chem. 275, 35715–35722 (2000).

    Article  CAS  Google Scholar 

  24. Gressner, A.M., Weiskirchen, R., Breitkopf, K. & Dooley, S. Roles of TGF-β in hepatic fibrosis. Front. Biosci. 7, d793–d807 (2002).

    Article  CAS  Google Scholar 

  25. Leask, A. & Abraham, D.J. TGF-β signaling and the fibrotic response. FASEB J. 18, 816–827 (2004).

    Article  CAS  Google Scholar 

  26. Breitling, R., Armengaud, P., Amtmann, A. & Herzyk, P. Rank products: a simple, yet powerful, new method to detect differentially regulated genes in replicated microarray experiments. FEBS Lett. 573, 83–92 (2004).

    Article  CAS  Google Scholar 

  27. Hong, F. et al. RankProd: a bioconductor package for detecting differentially expressed genes in meta-analysis. Bioinformatics 22, 2825–2827 (2006).

    Article  CAS  Google Scholar 

  28. Eden, S., Hashimshony, T., Keshet, I., Cedar, H. & Thorne, A.W. DNA methylation models histone acetylation. Nature 394, 842 (1998).

    Article  CAS  Google Scholar 

  29. Zilberman, D., Coleman-Derr, D., Ballinger, T. & Henikoff, S. Histone H2A.Z and DNA methylation are mutually antagonistic chromatin marks. Nature 456, 125–129 (2008).

    Article  CAS  Google Scholar 

  30. Kobor, M.S. & Lorincz, M.C. H2A.Z and DNA methylation: irreconcilable differences. Trends Biochem. Sci. 34, 158–161 (2009).

    Article  CAS  Google Scholar 

  31. Creyghton, M.P. et al. H2AZ is enriched at polycomb complex target genes in ES cells and is necessary for lineage commitment. Cell 135, 649–661 (2008).

    Article  CAS  Google Scholar 

  32. Gatewood, J.M., Cook, G.R., Balhorn, R., Schmid, C.W. & Bradbury, E.M. Isolation of four core histones from human sperm chromatin representing a minor subset of somatic histones. J. Biol. Chem. 265, 20662–20666 (1990).

    CAS  Google Scholar 

  33. Hammoud, S.S. et al. Distinctive chromatin in human sperm packages genes for embryo development. Nature 460, 473–478 (2009).

    Article  CAS  Google Scholar 

  34. Chou, S.T. & Gibson, J.B. A histochemical study of the bile ducts in long-term biliary obstruction in the rat. J. Pathol. 103, 163–175 (1971).

    Article  CAS  Google Scholar 

  35. De Minicis, S. et al. Gene expression profiles during hepatic stellate cell activation in culture and in vivo. Gastroenterology 132, 1937–1946 (2007).

    Article  CAS  Google Scholar 

  36. Anway, M.D., Cupp, A.S., Uzumcu, M. & Skinner, M.K. Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 308, 1466–1469 (2005).

    Article  CAS  Google Scholar 

  37. Ng, S.F. et al. Chronic high-fat diet in fathers programs β-cell dysfunction in female rat offspring. Nature 467, 963–966 (2010).

    Article  CAS  Google Scholar 

  38. Carone, B.R. et al. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell 143, 1084–1096 (2010).

    Article  CAS  Google Scholar 

  39. Gama-Sosa, M.A. et al. Tissue-specific differences in DNA methylation in various mammals. Biochim. Biophys. Acta 740, 212–219 (1983).

    Article  CAS  Google Scholar 

  40. Ghosh, S. et al. Tissue specific DNA methylation of CpG islands in normal human adult somatic tissues distinguishes neural from non-neural tissues. Epigenetics 5, 527–538 (2010).

    Article  CAS  Google Scholar 

  41. Nagase, H. & Ghosh, S. Epigenetics: differential DNA methylation in mammalian somatic tissues. FEBS J. 275, 1617–1623 (2008).

    Article  CAS  Google Scholar 

  42. McKay, J.A. et al. Blood as a surrogate marker for tissue-specific DNA methylation and changes due to folate depletion in post-partum female mice. Mol. Nutr. Food Res. 55, 1026–1035 (2011).

    Article  CAS  Google Scholar 

  43. Abdalla, H., Yoshizawa, Y. & Hochi, S. Active demethylation of paternal genome in mammalian zygotes. J. Reprod. Dev. 55, 356–360 (2009).

    Article  CAS  Google Scholar 

  44. Sasaki, H. & Matsui, Y. Epigenetic events in mammalian germ-cell development: reprogramming and beyond. Nat. Rev. Genet. 9, 129–140 (2008).

    Article  CAS  Google Scholar 

  45. Jaenisch, R. DNA methylation and imprinting: why bother? Trends Genet. 13, 323–329 (1997).

    Article  CAS  Google Scholar 

  46. Li, E., Beard, C. & Jaenisch, R. Role for DNA methylation in genomic imprinting. Nature 366, 362–365 (1993).

    Article  CAS  Google Scholar 

  47. Miller, D., Brinkworth, M. & Iles, D. Paternal DNA packaging in spermatozoa: more than the sum of its parts? DNA, histones, protamines and epigenetics. Reproduction 139, 287–301 (2010).

    Article  CAS  Google Scholar 

  48. Johnson, G.D. et al. The sperm nucleus: chromatin, RNA, and the nuclear matrix. Reproduction 141, 21–36 (2011).

    Article  CAS  Google Scholar 

  49. Jia, C. Advances in the regulation of liver regeneration. Expert Rev. Gastroenterol. Hepatol. 5, 105–121 (2011).

    Article  CAS  Google Scholar 

  50. Oakley, F. et al. Inhibition of inhibitor of κB kinases stimulates hepatic stellate cell apoptosis and accelerated recovery from rat liver fibrosis. Gastroenterology 128, 108–120 (2005).

    Article  CAS  Google Scholar 

  51. Oakley, F. et al. Nuclear factor-κB1 (p50) limits the inflammatory and fibrogenic responses to chronic injury. Am. J. Pathol. 166, 695–708 (2005).

    Article  CAS  Google Scholar 

  52. Gieling, R.G. et al. The c-Rel subunit of nuclear factor-κB regulates murine liver inflammation, wound-healing, and hepatocyte proliferation. Hepatology 51, 922–931 (2010).

    Article  CAS  Google Scholar 

  53. McKay, J.A., Wong, Y.K., Relton, C.L., Ford, D. & Mathers, J.C. Maternal folate supply and sex influence gene-specific DNA methylation in the fetal gut. Mol. Nutr. Food Res. 55, 1717–1723 (2011).

    Article  CAS  Google Scholar 

  54. Pittenger, M.F. et al. Multilineage potential of adult human mesenchymal stem cells. Science 284, 143–147 (1999).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Cheshire for technical assistance and N. Perkins for helpful suggestions with writing the manuscript, M. Bashton for bioinformatic support with microarray analysis and R. Kendall for assistance with rat studies. We also thank J. Kirby and J. Brain for their help with obtaining ethical approval for the retrospective patient study. This work was supported by grants from US National Institutes of Health National Institute of Alcohol Abuse and Alcoholism (to J.M. and D.A.M.) grant numbers 1U01AA018663-01 and R21AA016682; Wellcome Trust grant numbers WT086755MA (to D.A.M.) and WT074472MA (to A.M.E.) and Newcastle Biomedical Research Centre and National Institute for Health Research (to J.M.). The Centre for Brain Ageing and Vitality is funded through the Lifelong Health and Wellbeing cross council initiative by the Medical Research Council, the Biotechnology and Biological Sciences Research Council, the Engineering and Physical Sciences Research Council and the Economic and Social Research Council (to J.C.M.) and the Turkish Association For The Study Of The Liver (to M.Z.).

Author information

Authors and Affiliations

Authors

Contributions

M.Z. performed the majority of the laboratory-based experiments and the related data analyses. J.M., T.H., Y.K.W., J.C.M., A.G. and C.L.W. performed a portion of the laboratory experiments and the related data analyses. C.R.F. carried out the surgery and all the in vivo experiments relating to unilateral ureteral obstruction. M.J.B. obtained, cultured and phenotyped human mesenchymal stem cells. S.M. and Q.M.A. obtained archival human liver biopsy tissue and divided patients into groups based on their case history and disease severity. A.D.B. carried out all morphological analyses of tissue sections and scoring of patient liver pathology. A.M.E., F.O. and J.M. carried out all in vivo experiments. J.M. and D.A.M. designed the experiments and wrote the manuscript. All authors discussed the paper and commented on the manuscript.

Corresponding authors

Correspondence to Derek A Mann or Jelena Mann.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1 and 2 (PDF 2369 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Zeybel, M., Hardy, T., Wong, Y. et al. Multigenerational epigenetic adaptation of the hepatic wound-healing response. Nat Med 18, 1369–1377 (2012). https://doi.org/10.1038/nm.2893

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2893

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