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:

miR-145 and miR-143 regulate smooth muscle cell fate and plasticity

Abstract

MicroRNAs (miRNAs) are regulators of myriad cellular events, but evidence for a single miRNA that can efficiently differentiate multipotent stem cells into a specific lineage or regulate direct reprogramming of cells into an alternative cell fate has been elusive. Here we show that miR-145 and miR-143 are co-transcribed in multipotent murine cardiac progenitors before becoming localized to smooth muscle cells, including neural crest stem-cell-derived vascular smooth muscle cells. miR-145 and miR-143 were direct transcriptional targets of serum response factor, myocardin and Nkx2-5 (NK2 transcription factor related, locus 5) and were downregulated in injured or atherosclerotic vessels containing proliferating, less differentiated smooth muscle cells. miR-145 was necessary for myocardin-induced reprogramming of adult fibroblasts into smooth muscle cells and sufficient to induce differentiation of multipotent neural crest stem cells into vascular smooth muscle. Furthermore, miR-145 and miR-143 cooperatively targeted a network of transcription factors, including Klf4 (Kruppel-like factor 4), myocardin and Elk-1 (ELK1, member of ETS oncogene family), to promote differentiation and repress proliferation of smooth muscle cells. These findings demonstrate that miR-145 can direct the smooth muscle fate and that miR-145 and miR-143 function to regulate the quiescent versus proliferative phenotype of smooth muscle cells.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: miR-143 and miR-145 are cardiac-specific and smooth-muscle-specific miRNAs.
Figure 2: SRF and Nkx2-5 directly regulate cardiac and smooth muscle expression of miR-143 and miR-145.
Figure 3: miR-145 directs vascular smooth muscle cell fate.
Figure 4: miR-143 and miR-145 target a network of factors to promote VSMC differentiation and repress proliferation.
Figure 5: Model of miR-143 and miR-145 regulation of smooth muscle cell proliferation and differentiation.

Similar content being viewed by others

References

  1. Kloosterman, W. P. & Plasterk, R. H. The diverse functions of microRNAs in animal development and disease. Dev. Cell 11, 441–450 (2006)

    Article  CAS  Google Scholar 

  2. Calin, G. A. & Croce, C. M. MicroRNA signatures in human cancers. Nature Rev. Cancer 6, 857–866 (2006)

    Article  ADS  CAS  Google Scholar 

  3. Zhao, Y. & Srivastava, D. A developmental view of microRNA function. Trends Biochem. Sci. 32, 189–197 (2007)

    Article  CAS  Google Scholar 

  4. Bartel, D. P. MicroRNAs: target recognition and regulatory functions. Cell 136, 215–233 (2009)

    Article  CAS  Google Scholar 

  5. Rajewsky, N. MicroRNA target predictions in animals. Nature Genet. 38 (Suppl). S8–S13 (2006)

    Article  CAS  Google Scholar 

  6. Vasudevan, S., Tong, Y. & Steitz, J. A. Switching from repression to activation: microRNAs can up-regulate translation. Science 318, 1931–1934 (2007)

    Article  ADS  CAS  Google Scholar 

  7. Zhao, Y., Samal, E. & Srivastava, D. Serum response factor regulates a muscle-specific microRNA that targets Hand2 during cardiogenesis. Nature 436, 214–220 (2005)

    Article  ADS  CAS  Google Scholar 

  8. Kwon, C., Han, Z., Olson, E. N. & Srivastava, D. MicroRNA1 influences cardiac differentiation in Drosophila and regulates Notch signaling. Proc. Natl Acad. Sci. USA 102, 18986–18991 (2005)

    Article  ADS  CAS  Google Scholar 

  9. Chen, J. F. et al. The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nature Genet. 38, 228–233 (2006)

    Article  CAS  Google Scholar 

  10. Zhao, Y. et al. Dysregulation of cardiogenesis, cardiac conduction, and cell cycle in mice lacking miRNA-1–2. Cell 129, 303–317 (2007)

    Article  CAS  Google Scholar 

  11. Ivey, K. N. et al. MicroRNA regulation of cell lineages in mouse and human embryonic stem cells. Cell Stem Cell 2, 219–229 (2008)

    Article  CAS  Google Scholar 

  12. Srivastava, D. Making or breaking the heart: from lineage determination to morphogenesis. Cell 126, 1037–1048 (2006)

    Article  CAS  Google Scholar 

  13. Kattman, S. J., Huber, T. L. & Keller, G. M. Multipotent flk-1+ cardiovascular progenitor cells give rise to the cardiomyocyte, endothelial, and vascular smooth muscle lineages. Dev. Cell 11, 723–732 (2006)

    Article  CAS  Google Scholar 

  14. Le Douarin, N. M., Creuzet, S., Couly, G. & Dupin, E. Neural crest cell plasticity and its limits. Development 131, 4637–4650 (2004)

    Article  CAS  Google Scholar 

  15. Ross, R. The pathogenesis of atherosclerosis: A perspective for the 1990s. Nature 362, 801–809 (1993)

    Article  ADS  CAS  Google Scholar 

  16. Owens, G. K., Kumar, M. S. & Wamhoff, B. R. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol. Rev. 84, 767–801 (2004)

    Article  CAS  Google Scholar 

  17. Yoshida, T. & Owens, G. K. Molecular determinants of vascular smooth muscle cell diversity. Circ. Res. 96, 280–291 (2005)

    Article  CAS  Google Scholar 

  18. Wang, Y., Medvid, R., Melton, C., Jaenisch, R. & Blelloch, R. DGCR8 is essential for microRNA biogenesis and silencing of embryonic stem cell self-renewal. Nature Genet. 39, 380–385 (2007)

    Article  CAS  Google Scholar 

  19. Cai, C. L. et al. Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev. Cell 5, 877–889 (2003)

    Article  CAS  Google Scholar 

  20. Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev. Biol. 1, 4 (2001)

    Article  CAS  Google Scholar 

  21. Moretti, A. et al. Multipotent embryonic isl1+ progenitor cells lead to cardiac, smooth muscle, and endothelial cell diversification. Cell 127, 1151–1165 (2006)

    Article  CAS  Google Scholar 

  22. Wang, Z. et al. Myocardin and ternary complex factors compete for SRF to control smooth muscle gene expression. Nature 428, 185–189 (2004)

    Article  ADS  CAS  Google Scholar 

  23. Wang, D. et al. Activation of cardiac gene expression by myocardin, a transcriptional cofactor for serum response factor. Cell 105, 851–862 (2001)

    Article  CAS  Google Scholar 

  24. Chen, J., Kitchen, C. M., Streb, J. W. & Miano, J. M. Myocardin: a component of a molecular switch for smooth muscle differentiation. J. Mol. Cell. Cardiol. 34, 1345–1356 (2002)

    Article  CAS  Google Scholar 

  25. Long, X., Bell, R. D., Gerthoffer, W. T., Zlokovic, B. V. & Miano, J. M. Myocardin is sufficient for a smooth muscle-like contractile phenotype. Arterioscler. Thromb. Vasc. Biol. 28, 1505–1510 (2008)

    Article  CAS  Google Scholar 

  26. Chen, C. Y. & Schwartz, R. J. Recruitment of the tinman homolog Nkx-2.5 by serum response factor activates cardiac alpha-actin gene transcription. Mol. Cell. Biol. 16, 6372–6384 (1996)

    Article  CAS  Google Scholar 

  27. Ji, R. et al. MicroRNA expression signature and antisense-mediated depletion reveal an essential role of MicroRNA in vascular neointimal lesion formation. Circ. Res. 100, 1579–1588 (2007)

    Article  CAS  Google Scholar 

  28. Krutzfeldt, J. et al. Silencing of microRNAs in vivo with 'antagomirs'. Nature 438, 685–689 (2005)

    Article  ADS  Google Scholar 

  29. Maurer, J. et al. Establishment and controlled differentiation of neural crest stem cell lines using conditional transgenesis. Differentiation 75, 580–591 (2007)

    Article  CAS  Google Scholar 

  30. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007)

    Article  CAS  Google Scholar 

  31. Liu, Y. et al. Kruppel-like factor 4 abrogates myocardin-induced activation of smooth muscle gene expression. J. Biol. Chem. 280, 9719–9727 (2005)

    Article  CAS  Google Scholar 

  32. House, S. J. & Singer, H. A. CaMKII-delta isoform regulation of neointima formation after vascular injury. Arterioscler. Thromb. Vasc. Biol. 28, 441–447 (2008)

    Article  CAS  Google Scholar 

  33. Mishra-Gorur, K., Singer, H. A. & Castellot, J. J. Heparin inhibits phosphorylation and autonomous activity of Ca2+/calmodulin-dependent protein kinase II in vascular smooth muscle cells. Am. J. Pathol. 161, 1893–1901 (2002)

    Article  CAS  Google Scholar 

  34. Xu, N., Papagiannakopoulos, T., Pan, G., Thomson, J. A. & Kosik, K. S. MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses pluripotency in human embryonic stem cells. Cell 137, 647–658 (2009)

    Article  CAS  Google Scholar 

  35. Yamagishi, H. et al. Tbx1 is regulated by tissue-specific forkhead proteins through a common Sonic hedgehog-responsive enhancer. Genes Dev. 17, 269–281 (2003)

    Article  CAS  Google Scholar 

  36. Wang, Z., Wang, D. Z., Pipes, G. C. & Olson, E. N. Myocardin is a master regulator of smooth muscle gene expression. Proc. Natl Acad. Sci. USA 100, 7129–7134 (2003)

    Article  ADS  CAS  Google Scholar 

  37. Yamamoto, M. et al. The roles of protein kinase C beta I and beta II in vascular smooth muscle cell proliferation. Exp. Cell Res. 240, 349–358 (1998)

    Article  CAS  Google Scholar 

  38. Sinha, S. et al. Assessment of contractility of purified smooth muscle cells derived from embryonic stem cells. Stem Cells 24, 1678–1688 (2006)

    Article  Google Scholar 

  39. Obernosterer, G., Martinez, J. & Alenius, M. Locked nucleic acid-based in situ detection of microRNAs in mouse tissue sections. Nature Protocols 2, 1508–1514 (2007)

    Article  CAS  Google Scholar 

  40. Kruger, J. & Rehmsmeier, M. RNAhybrid: microRNA target prediction easy, fast and flexible. Nucleic Acids Res. 34, W451–W454 (2006)

    Article  Google Scholar 

  41. Zuker, M. Mfold web server for nucleic acid folding and hybridization prediction. Nucleic Acids Res. 31, 3406–3415 (2003)

    Article  CAS  Google Scholar 

  42. Regan, C. P., Adam, P. J., Madsen, C. S. & Owens, G. K. Molecular mechanisms of decreased smooth muscle differentiation marker expression after vascular injury. J. Clin. Invest. 106, 1139–1147 (2000)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank R. Blelloch for DGCR8-null EBs; R.J. Schwartz for SRF-null ES cells; I. Charo and N. Saederup for RNA from atherosclerotic tissue; J. Maurer for JoMa neural crest cell line; L. Qian and Y. Huang for providing mouse cardiac infarct RNA; C. Tsou for help with calcium flux assays; E. N. Olson for the myocardin expression plasmid; P. Swinton for generation of transgenic mice; J. Fish and C. Miller for histopathology support; S. Ordway and G. Howard for scientific editing; B. Taylor for manuscript preparation. We also thank members of the Srivastava laboratory for discussions. J.M.M. was supported by HL62572 and HL091168 from NHLBI/NIH. D.S. was supported by grants from the NHLBI/NIH and the California Institute for Regenerative Medicine (CIRM) and was an Established Investigator of the American Heart Association. This work was also supported by NIH/NCRR grant C06 RR018928 to the Gladstone Institutes.

Author Contributions K.R.C. and D.S. designed the study and K.R.C. executed or oversaw execution of all experiments; N.T.S. and E.C.B. performed the NCC studies; M.P.W. and K.N.I. performed some expression and stem cell studies and K.N.I. helped supervise the project; A.N.M. provided technical support; T.-H.L. and J.M.M. performed carotid artery ligation studies; S.U.M. isolated YFP+ progenitor cells and performed some expression studies; J.M.M. assisted K.R.C and D.S. in editing the manuscript; K.R.C. and D.S. wrote the manuscript and D.S. supervised all aspects of the project.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Deepak Srivastava.

Ethics declarations

Competing interests

D.S. serves on the Scientific Advisory Board of iZumi Bio.

Supplementary information

Supplementary Figures

This file contains Supplementary Figures 1-7 with Legends. Supplementary Fig. 3b was corrected on 18 August 2009. (PDF 1337 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Cordes, K., Sheehy, N., White, M. et al. miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature 460, 705–710 (2009). https://doi.org/10.1038/nature08195

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature08195

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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