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:

Methylation determines fibroblast activation and fibrogenesis in the kidney

Abstract

Fibrogenesis is a pathological wound repair process that fails to cease, even when the initial insult has been removed. Fibroblasts are principal mediators of fibrosis, and fibroblasts from fibrotic tissues fail to return to their quiescent stage, including when cultured in vitro. In a search for underlying molecular mechanisms, we hypothesized that this perpetuation of fibrogenesis is caused by epigenetic modifications. We demonstrate here that hypermethylation of RASAL1, encoding an inhibitor of the Ras oncoprotein, is associated with the perpetuation of fibroblast activation and fibrogenesis in the kidney. RASAL1 hypermethylation is mediated by the methyltransferase Dnmt1 in renal fibrogenesis, and kidney fibrosis is ameliorated in Dnmt1+/− heterozygous mice. These studies demonstrate that epigenetic modifications may provide a molecular basis for perpetuated fibroblast activation and fibrogenesis in the kidney.

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: 5′-azacytidine ameliorates experimental renal fibrosis.
Figure 2: RASAL1 hypermethylation in fibrotic kidney fibroblasts.
Figure 3: Absence of Rasal1 hypermethylation in kidneys that do not become fibrotic upon injury.
Figure 4: Rasal1 silencing causes renal fibroblast activation via Ras hyperactivity.
Figure 5: Dnmts in the mouse model of folic acid–induced nephropathy.
Figure 6: TGF-β1–induced methylation of Rasal1 in kidney fibroblasts is mediated by Dnmt1.

Similar content being viewed by others

References

  1. Eddy, A.A. Molecular insights into renal interstitial fibrosis. J. Am. Soc. Nephrol. 7, 2495–2508 (1996).

    CAS  PubMed  Google Scholar 

  2. Kuncio, G.S., Neilson, E.G. & Haverty, T. Mechanisms of tubulointerstitial fibrosis. Kidney Int. 39, 550–556 (1991).

    Article  CAS  PubMed  Google Scholar 

  3. Strutz, F. & Zeisberg, M. Renal fibroblasts and myofibroblasts in chronic kidney disease. J. Am. Soc. Nephrol. 17, 2992–2998 (2006).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  5. Müller, G.A. & Rodemann, H.P. Characterization of human renal fibroblasts in health and disease: I. Immunophenotyping of cultured tubular epithelial cells and fibroblasts derived from kidneys with histologically proven interstitial fibrosis. Am. J. Kidney Dis. 17, 680–683 (1991).

    Article  PubMed  Google Scholar 

  6. Rodemann, H.P. & Müller, G.A. Characterization of human renal fibroblasts in health and disease: II. In vitro growth, differentiation, and collagen synthesis of fibroblasts from kidneys with interstitial fibrosis. Am. J. Kidney Dis. 17, 684–686 (1991).

    Article  CAS  PubMed  Google Scholar 

  7. Weber, K.T. & Brilla, C.G. Factors associated with reactive and reparative fibrosis of the myocardium. Basic Res. Cardiol. 87, Suppl 1, 291–301 (1992).

    PubMed  Google Scholar 

  8. Hinz, B. et al. The myofibroblast: one function, multiple origins. Am. J. Pathol. 170, 1807–1816 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lafyatis, R. Targeting fibrosis in systemic sclerosis. Endocr. Metab. Immune Disord. Drug Targets 6, 395–400 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Lin, J. et al. Kielin/chordin-like protein, a novel enhancer of BMP signaling, attenuates renal fibrotic disease. Nat. Med. 11, 387–393 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. Santini, V., Kantarjian, H.M. & Issa, J.P. Changes in DNA methylation in neoplasia: pathophysiology and therapeutic implications. Ann. Intern. Med. 134, 573–586 (2001).

    Article  CAS  PubMed  Google Scholar 

  12. Bird, A.P. & Wolffe, A.P. Methylation-induced repression—belts, braces, and chromatin. Cell 99, 451–454 (1999).

    Article  CAS  PubMed  Google Scholar 

  13. Feinberg, A.P. & Vogelstein, B. Alterations in DNA methylation in human colon neoplasia. Semin. Surg. Oncol. 3, 149–151 (1987).

    Article  CAS  PubMed  Google Scholar 

  14. Herman, J.G. & Baylin, S.B. Gene silencing in cancer in association with promoter hypermethylation. N. Engl. J. Med. 349, 2042–2054 (2003).

    Article  CAS  PubMed  Google Scholar 

  15. Jones, P.A. & Baylin, S.B. The epigenomics of cancer. Cell 128, 683–692 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Clark, S.J., Statham, A., Stirzaker, C., Molloy, P.L. & Frommer, M. DNA methylation: bisulphite modification and analysis. Nat. Protoc. 1, 2353–2364 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Gius, D. et al. The epigenome as a molecular marker and target. Cancer 104, 1789–1793 (2005).

    Article  CAS  PubMed  Google Scholar 

  18. Downward, J. Targeting RAS signalling pathways in cancer therapy. Nat. Rev. Cancer 3, 11–22 (2003).

    Article  CAS  PubMed  Google Scholar 

  19. Mitin, N., Rossman, K.L. & Der, C.J. Signaling interplay in Ras superfamily function. Curr. Biol. 15, R563–R574 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Walker, S.A. et al. Identification of a Ras GTPase-activating protein regulated by receptor-mediated Ca2+ oscillations. EMBO J. 23, 1749–1760 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Barbacid, M. Ras genes. Annu. Rev. Biochem. 56, 779–827 (1987).

    Article  CAS  PubMed  Google Scholar 

  22. Bos, J.L. Ras oncogenes in human cancer: a review. Cancer Res. 49, 4682–4689 (1989).

    CAS  PubMed  Google Scholar 

  23. Arun, D. & Gutmann, D.H. Recent advances in neurofibromatosis type 1. Curr. Opin. Neurol. 17, 101–105 (2004).

    Article  CAS  PubMed  Google Scholar 

  24. Kolfschoten, I.G. et al. A genetic screen identifies PITX1 as a suppressor of RAS activity and tumorigenicity. Cell 121, 849–858 (2005).

    Article  CAS  PubMed  Google Scholar 

  25. Jin, H. et al. Epigenetic silencing of a Ca2+-regulated Ras GTPase-activating protein RASAL defines a new mechanism of Ras activation in human cancers. Proc. Natl. Acad. Sci. USA 104, 12353–12358 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Gazin, C., Wajapeyee, N., Gobeil, S., Virbasius, C.M. & Green, M.R. An elaborate pathway required for Ras-mediated epigenetic silencing. Nature 449, 1073–1077 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lloyd, C.M. et al. RANTES and monocyte chemoattractant protein-1 (MCP-1) play an important role in the inflammatory phase of crescentic nephritis, but only MCP-1 is involved in crescent formation and interstitial fibrosis. J. Exp. Med. 185, 1371–1380 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zeisberg, M. et al. BMP-7 counteracts TGF-β1–induced epithelial-to-mesenchymal transition and reverses chronic renal injury. Nat. Med. 9, 964–968 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Witzgall, R., Brown, D., Schwarz, C. & Bonventre, J.V. Localization of proliferating cell nuclear antigen, vimentin, c-Fos and clusterin in the postischemic kidney. Evidence for a heterogenous genetic response among nephron segments, and a large pool of mitotically active and dedifferentiated cells. J. Clin. Invest. 93, 2175–2188 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Oliver, J.A., Maarouf, O., Cheema, F.H., Martens, T.P. & Al-Awqati, Q. The renal papilla is a niche for adult kidney stem cells. J. Clin. Invest. 114, 795–804 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hendry, B.M. & Sharpe, C.C. Targeting Ras genes in kidney disease. Nephron 93, e129–e133 (2003).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  33. Okano, M., Bell, D.W., Haber, D.A. & Li, E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99, 247–257 (1999).

    Article  CAS  PubMed  Google Scholar 

  34. Li, E., Bestor, T.H. & Jaenisch, R. Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69, 915–926 (1992).

    Article  CAS  PubMed  Google Scholar 

  35. Bottinger, E.P. & Bitzer, M. TGF-β signaling in renal disease. J. Am. Soc. Nephrol. 13, 2600–2610 (2002).

    Article  PubMed  Google Scholar 

  36. Border, W.A. & Noble, N.A. Targeting TGF-β for treatment of disease. Nat. Med. 1, 1000–1001 (1995).

    Article  CAS  PubMed  Google Scholar 

  37. Strutz, F. et al. TGF-β1 induces proliferation in human renal fibroblasts via induction of basic fibroblast growth factor (FGF-2). Kidney Int. 59, 579–592 (2001).

    Article  CAS  PubMed  Google Scholar 

  38. Basile, D.P. et al. Identification of persistently altered gene expression in the kidney after functional recovery from ischemic acute renal failure. Am. J. Physiol. Renal Physiol. 288, F953–F963 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. Forbes, J.M., Leaker, B., Hewitson, T.D., Becker, G.J. & Jones, C.L. Macrophage and myofibroblast involvement in ischemic acute renal failure is attenuated by endothelin receptor antagonists. Kidney Int. 55, 198–208 (1999).

    Article  CAS  PubMed  Google Scholar 

  40. Villanueva, S., Cespedes, C. & Vio, C.P. Ischemic acute renal failure induces the expression of a wide range of nephrogenic proteins. Am. J. Physiol. Regul. Integr. Comp. Physiol. 290, R861–R870 (2006).

    Article  CAS  PubMed  Google Scholar 

  41. Ding, M. et al. Loss of the tumor suppressor Vhlh leads to upregulation of Cxcr4 and rapidly progressive glomerulonephritis in mice. Nat. Med. 12, 1081–1087 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Rothenpieler, U.W. & Dressler, G.R. Pax-2 is required for mesenchyme-to-epithelium conversion during kidney development. Development 119, 711–720 (1993).

    CAS  PubMed  Google Scholar 

  43. Khwaja, A., Sharpe, C.C., Noor, M., Kloog, Y. & Hendry, B.M. The inhibition of human mesangial cell proliferation by S-trans, trans-farnesylthiosalicylic acid. Kidney Int. 68, 474–486 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Zaidi, N. et al. A new approach for distinguishing cathepsin E and D activity in antigen-processing organelles. FEBS J. 274, 3138–3149 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Zeisberg, E.M. et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat. Med. 13, 952–961 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Zeisberg, E.M., Potenta, S.E., Sugimoto, H., Zeisberg, M. & Kalluri, R. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J. Am. Soc. Nephrol. 19, 2282–2287 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Chatterjee, P.K. et al. Calpain inhibitor-1 reduces renal ischemia/reperfusion injury in the rat. Kidney Int. 59, 2073–2083 (2001).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This study was partially funded by research grants R01DK081576 (M.Z.), R03DK081687 (M.Z.), R01DK30932 (D.J.S.), R01DK55001 (R.K.), R01CA125550 (R.K.), Mentored Clinical Scientist Development Awards 1K08 CA129204 (E.M.Z.) and K08 DK074558 (M.Z.) from the National Institutes of Health, the American Society of Nephrology Carl W. Gottschalk Scholar Grant (M.Z.), American Heart Association Scientist Development Grant SDG0735602T, Deutsche Forschungsgemeinschaft-Stipendium BE4211/1-1 (W.B.), Champalimaud Metastasis Research Program (R.K.) and research funds from the Beth Israel Deaconess Medical Center for the Division of Matrix Biology. We thank A. Lau and T. Zhong Hu for technical assistance. FSP-1–specific antibodies were a gift from E. Neilson (Vanderbilt University).

Author information

Authors and Affiliations

Authors

Contributions

W.B. performed and designed experiments, analyzed data and edited the manuscript. S.M. performed experiments and analyzed data. E.M.Z. advised, performed experiments, analyzed data and edited the manuscript. G.A.M. and C.A.M. characterized and provided human fibroblasts. H.K. generated Rasal1 antibodies. D.J.S. provided nephrotoxic serum and edited the manuscript. R.K. advised and edited the manuscript. M.Z. designed, performed and supervised experiments, analyzed data and wrote the manuscript.

Corresponding author

Correspondence to Michael Zeisberg.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 and Supplementary Tables 1–3 (PDF 2376 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bechtel, W., McGoohan, S., Zeisberg, E. et al. Methylation determines fibroblast activation and fibrogenesis in the kidney. Nat Med 16, 544–550 (2010). https://doi.org/10.1038/nm.2135

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2135

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