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:

Snail1-induced partial epithelial-to-mesenchymal transition drives renal fibrosis in mice and can be targeted to reverse established disease

An Erratum to this article was published on 04 February 2016

This article has been updated

Abstract

Progressive kidney fibrosis contributes greatly to end-stage renal failure, and no specific treatment is available to preserve organ function. During renal fibrosis, myofibroblasts accumulate in the interstitium of the kidney, leading to massive deposition of extracellular matrix and organ dysfunction. The origin of myofibroblasts is manifold, but the contribution of an epithelial-to-mesenchymal transition (EMT) undergone by renal epithelial cells during kidney fibrosis is still debated. We show that the reactivation of Snai1 (encoding snail family zinc finger 1, known as Snail1) in mouse renal epithelial cells is required for the development of fibrosis in the kidney. Damage-mediated Snail1 reactivation induces a partial EMT in tubular epithelial cells that, without directly contributing to the myofibroblast population, relays signals to the interstitium to promote myofibroblast differentiation and fibrogenesis and to sustain inflammation. We also show that Snail1-induced fibrosis can be reversed in vivo and that obstructive nephropathy can be therapeutically ameliorated in mice by targeting Snail1 expression. These results reconcile conflicting data on the role of the EMT in renal fibrosis and provide avenues for the design of novel anti-fibrotic therapies.

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: Snail1 reactivation is required for the development of UUO-induced fibrosis.
Figure 2: Renal epithelial cells undergo a partial EMT after UUO.
Figure 3: Snail1 reactivation in renal epithelial cells is required for sustained inflammation in the injured kidney.
Figure 4: Snail1 reactivation is required for the development of folic acid-induced fibrosis.
Figure 5: Snail1-induced fibrosis can be reversed in vivo.
Figure 6: Snail1 inactivation reverts UUO-induced fibrosis in mice.

Similar content being viewed by others

Change history

  • 04 September 2015

    In the version of this article initially published, the word 'genetically' was added by the editor to the penultimate sentence of the abstract during proofing of the manuscript. However, the authors used not only a genetic knockout approach but also morpholino-induced inhibition of proper mRNA processing to target Snail1 expression. Thus, the word 'genetically' has been deleted from the sentence to better convey the findings of the report. The error has been corrected in the HTML and PDF versions of the article.

References

  1. Liu, Y. Cellular and molecular mechanisms of renal fibrosis. Nat. Rev. Nephrol. 7, 684–696 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. National Institute of Diabetes and Digestive and Kidney Diseases. Kidney Disease Statistics for the United States. No. 12–3895, http://www.niddk.nih.gov/health-information/health-statistics/Pages/kidney-disease-statistics-united-states.aspx#1 (2012).

  3. Dressler, G. Tubulogenesis in the developing mammalian kidney. Trends Cell Biol. 12, 390–395 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Thiery, J.P., Acloque, H., Huang, R.Y. & Nieto, M.A. Epithelial–mesenchymal transitions in development and disease. Cell 139, 871–890 (2009).

    Article  CAS  PubMed  Google Scholar 

  5. Boutet, A. et al. Snail activation disrupts tissue homeostasis and induces fibrosis in the adult kidney. EMBO J. 25, 5603–5613 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sato, M., Muragaki, Y., Saika, S., Roberts, A.B. & Ooshima, A. Targeted disruption of TGF-beta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J. Clin. Invest. 112, 1486–1494 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lange-Sperandio, B. et al. Leukocytes induce epithelial to mesenchymal transition after unilateral ureteral obstruction in neonatal mice. Am. J. Pathol. 171, 861–871 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Grande, M.T. et al. Deletion of H-Ras decreases renal fibrosis and myofibroblast activation following ureteral obstruction in mice. Kidney Int. 77, 509–518 (2010).

    Article  CAS  PubMed  Google Scholar 

  9. Chevalier, R.L., Forbes, M.S. & Thornhill, B.A. Ureteral obstruction as a model of renal interstitial fibrosis and obstructive nephropathy. Kidney Int. 75, 1145–1152 (2009).

    Article  PubMed  Google Scholar 

  10. Iwano, M. et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J. Clin. Invest. 110, 341–350 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Humphreys, B.D. et al. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am. J. Pathol. 176, 85–97 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Li, L., Zepeda-Orozco, D., Black, R. & Lin, F. Autophagy is a component of epithelial cell fate in obstructive uropathy. Am. J. Pathol. 176, 1767–1778 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. LeBleu, V.S. et al. Origin and function of myofibroblasts in kidney fibrosis. Nat. Med. 19, 1047–1053 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Rowe, R.G. et al. Mesenchymal cells reactivate Snail1 expression to drive three-dimensional invasion programs. J. Cell Biol. 184, 399–408 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Shao, X., Somlo, S. & Igarashi, P. Epithelial-specific Cre/lox recombination in the developing kidney and genitourinary tract. J. Am. Soc. Nephrol. 13, 1837–1846 (2002).

    Article  CAS  PubMed  Google Scholar 

  16. Franci, C. et al. Expression of Snail in tumor-stroma interface. Oncogene 25, 5134–5144 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. de Oliveira Costa, M.Z., Bacchi, C.E. & Franco, M. Histogenesis of the acquired cystic kidney disease: an immunohistochemical study. Appl. Immunohistochem. Mol. Morphol. 14, 348–352 (2006).

    Article  Google Scholar 

  18. Borges, F.T. et al. TGF-β-1-containing exosomes from injured epithelial cells activate fibroblasts to initiate tissue regenerative responses and fibrosis. J. Am. Soc. Nephrol. 24, 385–392 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Dhasarathy, A., Phadke, D., Mav, D., Shah, R.R. & Wade, P.A. The transcription factors Snail and Slug activate the transforming growth factor-β signaling pathway in breast cancer. PLoS ONE 6, e26514 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Grande, M.T., Pérez-Barriocanal, F. & López-Novoa, J.M. Role of inflammation in tubulo-interstitial damage associated to obstructive nephropathy. J. Inflamm. (Lond.) 7, 19 (2010).

    Article  CAS  Google Scholar 

  21. Biswas, S.K. & Mantovani, A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat. Immunol. 11, 889–896 (2010).

    CAS  PubMed  Google Scholar 

  22. Lyons, J.G. et al. Snail upregulates pro-inflammatory mediators and inhibits differentiation in oral keratinocytes. Cancer Res. 68, 4525–4530 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hsu, D.S. et al. Acetylation of Snail modulates the cytokinome of cancer cells to enhance the recruitment of macrophages. Cancer Cell 26, 534–548 (2014).

    Article  CAS  PubMed  Google Scholar 

  24. Yang, H.-C., Zuo, Y. & Fogo, A.B. Models of chronic disease. Drug Discov. Today Dis. Models 7, 13–19 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Nieto, M.A. Epithelial plasticity: a common theme in embryonic and cancer cells. Science 342, 1234850 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Kriz, W., Kaissling, B. & Le Hir, M. Epithelial–mesenchymal transition (EMT) in kidney fibrosis: fact or fantasy? J. Clin. Invest. 121, 468–474 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Fragiadaki, M. & Mason, R.M. Epithelial–mesenchymal transition in renal fibrosis - evidence for and against. Int. J. Exp. Pathol. 92, 143–150 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zeisberg, M. & Neilson, E.G. Mechanisms of tubulointerstitial fibrosis. J. Am. Soc. Nephrol. 21, 1819–1834 (2010).

    Article  CAS  PubMed  Google Scholar 

  29. Meng, X.M., Nikolic-Paterson, D.J. & Lan, H.Y. Inflammatory processes in renal fibrosis. Nat. Rev. Nephrol. 10, 493–503 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Vega, S. et al. Snail blocks the cell cycle and confers resistance to cell death. Genes Dev. 18, 1131–1143 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kim, N.H. et al. A p53/miRNA-34 axis regulates Snail1-dependent cancer cell epithelial-mesenchymal transition. J. Cell Biol. 195, 417–433 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Tian, X.J., Zhang, H. & Xing, J. Coupled reversible and irreversible bistable switches underlying TGFbeta-induced epithelial to mesenchymal transition. Biophys. J. 105, 1079–1089 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Morizane, R. et al. miR-34c attenuates epithelial-mesenchymal transition and kidney fibrosis with ureteral obstruction. Sci. Rep. 4, 4578 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kalluri, R. & Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Invest. 119, 1420–1428 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Liu, Y. Renal fibrosis: new insights into the pathogenesis and therapeutics. Kidney Int. 69, 213–217 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. López-Novoa, J.M. & Nieto, M.A. Inflammation and EMT: an alliance towards organ fibrosis and cancer progression. EMBO Mol. Med. 1, 303–314 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wu, Y. et al. Stabilization of snail by NF-kappaB is required for inflammation-induced cell migration and invasion. Cancer Cell 15, 416–428 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Rosenbloom, J., Castro, S.V. & Jimenez, S.A. Narrative review: fibrotic diseases: cellular and molecular mechanisms and novel therapies. Ann. Intern. Med. 152, 159–166 (2010).

    Article  PubMed  Google Scholar 

  39. 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 

  40. Esteban, V. et al. Angiotensin II, via AT1 and AT2 receptors and NF-kappaB pathway, regulates the inflammatory response in unilateral ureteral obstruction. J. Am. Soc. Nephrol. 15, 1514–1529 (2004).

    Article  CAS  PubMed  Google Scholar 

  41. Miyajima, A. et al. Novel nuclear factor kappa B activation inhibitor prevents inflammatory injury in unilateral ureteral obstruction. J. Urol. 169, 1559–1563 (2003).

    Article  CAS  PubMed  Google Scholar 

  42. Mehal, W.Z., Iredale, J. & Friedman, S.L. Scraping fibrosis: expressway to the core of fibrosis. Nat. Med. 17, 552–553 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Luedde, T. & Schwabe, R.F. NF-κB in the liver–linking injury, fibrosis and hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 8, 108–118 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Tsai, J.H., Donaher, J.L., Murphy, D.A., Chau, S. & Yang, J. Spatiotemporal regulation of epithelial-mesenchymal transition is essential for squamous cell carcinoma metastasis. Cancer Cell 22, 725–736 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ocaña, O.H. et al. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell 22, 709–724 (2012).

    Article  CAS  PubMed  Google Scholar 

  46. Rodríguez-López, A., Flores, O., Arévalo, M. & López-Novoa, J.M. Glomerular cell proliferation and apoptosis in the early phase of renal damage in uninephrectomized spontaneously hypertensive rats. Kidney Int. 54, S36–S40 (1998).

    Article  Google Scholar 

Download references

Acknowledgements

We thank members of A. Nieto's laboratory for their helpful discussions and comments throughout the years. We also thank S. Canals for his advice on statistical analysis, and C. Villena and S. Ingham for the design of Supplementary Figure 10. We are very grateful to P. Igarashi (University of Minnesota Medical School) for providing the Ksp1.3-Cre mice. This work was supported by grants from the Spanish Ministry of Economy and Competitiveness (BFU2008-01042 and CONSOLIDER-INGENIO 2010 CSD2007-00023 and CDS2007-00017), the Generalitat Valenciana (Prometeo 2008/049 and PROMETEOII/2013/002) and the European Research Council (ERC AdG 322694) to M.A.N. The Instituto de Neurociencias is a Centre of Excellence Severo Ochoa. M.T.G. was a recipient of a contract from the Junta de Ampliación de Estudios program at the Consejo Superior de Investigaciones Científicas European Social Fund.

Author information

Authors and Affiliations

Authors

Contributions

M.T.G. and B.S.-L. performed the majority of experiments, analyzed the data and contributed to writing the manuscript. C.L.-B. was instrumental in the surgery, and histological and expression studies. C.A.F. and A.B. started the project, R.G.R. and S.J.W. provided the Snai1loxP mice before publication, J.M.L.-N. and M.A. quantified Sirius red staining and J.M.L.-N. helped in the interpretation of data. M.A.N. conceived the project, interpreted the data and wrote the manuscript.

Corresponding author

Correspondence to M Angela Nieto.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 & Supplementary Table 1 (PDF 13042 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Grande, M., Sánchez-Laorden, B., López-Blau, C. et al. Snail1-induced partial epithelial-to-mesenchymal transition drives renal fibrosis in mice and can be targeted to reverse established disease. Nat Med 21, 989–997 (2015). https://doi.org/10.1038/nm.3901

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3901

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