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.

  • Letter
  • Published:

LIN28 phosphorylation by MAPK/ERK couples signalling to the post-transcriptional control of pluripotency

Abstract

Signalling and post-transcriptional gene control are both critical for the regulation of pluripotency1,2, yet how they are integrated to influence cell identity remains poorly understood. LIN28 (also known as LIN28A), a highly conserved RNA-binding protein, has emerged as a central post-transcriptional regulator of cell fate through blockade of let-7 microRNA biogenesis and direct modulation of mRNA translation3. Here we show that LIN28 is phosphorylated by MAPK/ERK in pluripotent stem cells, which increases its levels via post-translational stabilization. LIN28 phosphorylation had little impact on let-7 but enhanced the effect of LIN28 on its direct mRNA targets, revealing a mechanism that uncouples LIN28’s let-7-dependent and -independent activities. We have linked this mechanism to the induction of pluripotency by somatic cell reprogramming and the transition from naive to primed pluripotency. Collectively, our findings indicate that MAPK/ERK directly impacts LIN28, defining an axis that connects signalling, post-transcriptional gene control, and cell fate regulation.

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: MAPK/ERK phosphorylates LIN28A on Ser200.
Figure 2: LIN28A phosphorylation increases its protein stability.
Figure 3: LIN28A phosphorylation can uncouple its let-7-dependent and -independent activities.
Figure 4: LIN28A phosphorylation contributes to the regulation of pluripotency transitions.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Ng, H. H. & Surani, M. A. The transcriptional and signalling networks of pluripotency. Nat. Cell Biol. 13, 490–496 (2011).

    Article  CAS  Google Scholar 

  2. Ye, J. & Blelloch, R. Regulation of pluripotency by RNA binding proteins. Cell Stem Cell 15, 271–280 (2014).

    Article  CAS  Google Scholar 

  3. Shyh-Chang, N. & Daley, G. Q. Lin28: primal regulator of growth and metabolism in stem cells. Cell Stem Cell 12, 395–406 (2013).

    Article  Google Scholar 

  4. Kunath, T. et al. FGF stimulation of the Erk1/2 signalling cascade triggers transition of pluripotent embryonic stem cells from self-renewal to lineage commitment. Development 134, 2895–2902 (2007).

    Article  CAS  Google Scholar 

  5. Ying, Q. L. et al. The ground state of embryonic stem cell self-renewal. Nature 453, 519–523 (2008).

    Article  CAS  Google Scholar 

  6. Moss, E. G., Lee, R. C. & Ambros, V. The cold shock domain protein LIN-28 controls developmental timing in C. elegans and is regulated by the lin-4 RNA. Cell 88, 637–646 (1997).

    Article  CAS  Google Scholar 

  7. Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917–1920 (2007).

    Article  CAS  Google Scholar 

  8. Kumar, R. M. et al. Deconstructing transcriptional heterogeneity in pluripotent stem cells. Nature 516, 56–61 (2014).

    Article  CAS  Google Scholar 

  9. Zhang, J. et al. LIN28 regulates stem cell metabolism and conversion to primed pluripotency. Cell Stem Cell 19, 66–80 (2016).

    Article  CAS  Google Scholar 

  10. Heo, I. et al. Lin28 mediates the terminal uridylation of let-7 precursor microRNA. Mol. Cell 32, 276–284 (2008).

    Article  CAS  Google Scholar 

  11. Newman, M. A., Thomson, J. M. & Hammond, S. M. Lin-28 interaction with the Let-7 precursor loop mediates regulated microRNA processing. RNA 14, 1539–1549 (2008).

    Article  CAS  Google Scholar 

  12. Rybak, A. et al. A feedback loop comprising lin-28 and let-7 controls pre-let-7 maturation during neural stem-cell commitment. Nat. Cell Biol. 10, 987–993 (2008).

    Article  CAS  Google Scholar 

  13. Viswanathan, S. R., Daley, G. Q. & Gregory, R. I. Selective blockade of microRNA processing by Lin28. Science 320, 97–100 (2008).

    Article  CAS  Google Scholar 

  14. Cho, J. et al. LIN28A is a suppressor of ER-associated translation in embryonic stem cells. Cell 151, 765–777 (2012).

    Article  CAS  Google Scholar 

  15. Peng, S. et al. Genome-wide studies reveal that Lin28 enhances the translation of genes important for growth and survival of human embryonic stem cells. Stem Cells 29, 496–504 (2011).

    Article  CAS  Google Scholar 

  16. Polesskaya, A. et al. Lin-28 binds IGF-2 mRNA and participates in skeletal myogenesis by increasing translation efficiency. Genes Dev. 21, 1125–1138 (2007).

    Article  CAS  Google Scholar 

  17. Shyh-Chang, N. et al. Lin28 enhances tissue repair by reprogramming cellular metabolism. Cell 155, 778–792 (2013).

    Article  CAS  Google Scholar 

  18. Wilbert, M. L. et al. LIN28 binds messenger RNAs at GGAGA motifs and regulates splicing factor abundance. Mol. Cell 48, 195–206 (2012).

    Article  CAS  Google Scholar 

  19. Rigbolt, K. T. et al. System-wide temporal characterization of the proteome and phosphoproteome of human embryonic stem cell differentiation. Sci. Signal. 4, rs3 (2011).

    Article  Google Scholar 

  20. Van Hoof, D. et al. Phosphorylation dynamics during early differentiation of human embryonic stem cells. Cell Stem Cell 5, 214–226 (2009).

    Article  CAS  Google Scholar 

  21. Mansour, S. J. et al. Transformation of mammalian cells by constitutively active MAP kinase kinase. Science 265, 966–970 (1994).

    Article  CAS  Google Scholar 

  22. Cai, N., Li, M., Qu, J., Liu, G. H. & Izpisua Belmonte, J. C. Post-translational modulation of pluripotency. J. Mol. Cell Biol. 4, 262–265 (2012).

    Article  CAS  Google Scholar 

  23. Dangi-Garimella, S. et al. Raf kinase inhibitory protein suppresses a metastasis signalling cascade involving LIN28 and let-7. EMBO J. 28, 347–358 (2009).

    Article  CAS  Google Scholar 

  24. Kwon, S. C. et al. The RNA-binding protein repertoire of embryonic stem cells. Nat. Struct. Mol. Biol. 20, 1122–1130 (2013).

    Article  CAS  Google Scholar 

  25. Tan, F. E. & Elowitz, M. B. Brf1 posttranscriptionally regulates pluripotency and differentiation responses downstream of Erk MAP kinase. Proc. Natl Acad. Sci. USA 111, E1740–E1748 (2014).

    Article  CAS  Google Scholar 

  26. Wang, L. X., Wang, J., Qu, T. T., Zhang, Y. & Shen, Y. F. Reversible acetylation of Lin28 mediated by PCAF and SIRT1. Biochim. Biophys. Acta 1843, 1188–1195 (2014).

    Article  CAS  Google Scholar 

  27. Kim, S. K. et al. SET7/9 methylation of the pluripotency factor LIN28A is a nucleolar localization mechanism that blocks let-7 biogenesis in human ESCs. Cell Stem Cell 15, 735–749 (2014).

    Article  CAS  Google Scholar 

  28. Balzer, E., Heine, C., Jiang, Q., Lee, V. M. & Moss, E. G. LIN28 alters cell fate succession and acts independently of the let-7 microRNA during neurogliogenesis in vitro. Development 137, 891–900 (2010).

    Article  CAS  Google Scholar 

  29. Vadla, B., Kemper, K., Alaimo, J., Heine, C. & Moss, E. G. lin-28 controls the succession of cell fate choices via two distinct activities. PLoS Genet. 8, e1002588 (2012).

    Article  CAS  Google Scholar 

  30. Morgenstern, J. P. & Land, H. Advanced mammalian gene transfer: high titre retroviral vectors with multiple drug selection markers and a complementary helper-free packaging cell line. Nucleic Acids Res. 18, 3587–3596 (1990).

    Article  CAS  Google Scholar 

  31. Zhu, H. et al. Lin28a transgenic mice manifest size and puberty phenotypes identified in human genetic association studies. Nat. Genet. 42, 626–630 (2010).

    Article  CAS  Google Scholar 

  32. Eggan, K. et al. Hybrid vigor, fetal overgrowth, and viability of mice derived by nuclear cloning and tetraploid embryo complementation. Proc. Natl Acad. Sci. USA 98, 6209–6214 (2001).

    Article  CAS  Google Scholar 

  33. Shinoda, G. et al. Fetal deficiency of lin28 programs life-long aberrations in growth and glucose metabolism. Stem Cells 31, 1563–1573 (2013).

    Article  CAS  Google Scholar 

  34. Park, I. H. et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature 451, 141–146 (2008).

    Article  CAS  Google Scholar 

  35. Khandelia, P., Yap, K. & Makeyev, E. V. Streamlined platform for short hairpin RNA interference and transgenesis in cultured mammalian cells. Proc. Natl Acad. Sci. USA 108, 12799–12804 (2011).

    Article  CAS  Google Scholar 

  36. Triboulet, R., Pirouz, M. & Gregory, R. I. A single let-7 microRNA bypasses LIN28-mediated repression. Cell Rep. 13, 260–266 (2015).

    Article  CAS  Google Scholar 

  37. Villen, J. & Gygi, S. P. The SCX/IMAC enrichment approach for global phosphorylation analysis by mass spectrometry. Nat. Protoc. 3, 1630–1638 (2008).

    Article  Google Scholar 

  38. Beausoleil, S. A., Villen, J., Gerber, S. A., Rush, J. & Gygi, S. P. A probability-based approach for high-throughput protein phosphorylation analysis and site localization. Nat. Biotechnol. 24, 1285–1292 (2006).

    Article  CAS  Google Scholar 

  39. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).

    Article  CAS  Google Scholar 

  40. Anders, S., Pyl, P. T. & Huber, W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank P. Sharp, L. Cantley, G. Ruvkun, and members of the Daley laboratory for invaluable discussions, A. D. L. Angeles for critical feedback on the manuscript, X. Wu/Yi Zhang’s laboratory and R. Rubio/DFCI CCCB for assistance with RNA-seq, and R. Tomaino at the Taplin Biological Mass Spectrometry Core for assistance with mass spectrometry. Bioanalyser analysis was performed in the BCH IDDRC Molecular Genetics Core, which is supported by NIH (NIH-P30-HD 18655). Sequencing analysis was conducted on the Orchestra High Performance Computing Cluster at Harvard Medical School. K.M.T. was an HHMI International Student Research Fellow and a Herchel Smith Graduate Fellow. D.S.P. was supported by a grant from NIGMS (T32GM007753). R.I.G. was supported by a grant from NIGMS (R01GM086386). G.Q.D. is an investigator of the Howard Hughes Medical Institute and the Manton Center for Orphan Disease Research, and was supported by a grant from NIGMS (R01GM107536).

Author information

Authors and Affiliations

Authors

Contributions

K.M.T. designed and performed the experiments, and wrote the manuscript. D.S.P. helped with RNA-seq, HeLa clonal series generation and expression analysis. Z.W. performed reprogramming experiments. A.H. performed RNA-seq bioinformatics analysis. R.T. and R.I.G. shared unpublished results and generated the isogenic HeLa cells. M.T.S. performed expression analysis. J.T.P. and J.K.O. helped with experimental design. S.K. generated the human pLIN28A antibody. S.P.G. supervised the proteomics experiments. G.Q.D. designed and supervised experiments, and wrote the manuscript.

Corresponding author

Correspondence to George Q. Daley.

Ethics declarations

Competing interests

G.Q.D. and R.I.G. hold options and intellectual property related to 28/7 Therapeutics, a company seeking to develop inhibitors of the LIN28/let-7 pathway. S.K. is an employee of Cell Signaling Technology.

Integrated supplementary information

Supplementary Figure 1 Phosphoproteomic analysis of LIN28A.

(a) Schematic of the phosphoproteomic strategy. (b) Coomassie-stained SDS-PAGE gel showing FLAG-LIN28A purified from mESCs, without or with Calyculin A (100 nM) treatment. (c) Digestion patterns of proteases used for generation of LIN28A peptides. The amino acid sequence of mouse LIN28A is shown. Trypsin and chymotrypsin sites are highlighted in blue and red, respectively. (d) LIN28A phosphosites and kinases predicted to phosphorylate them, as determined by ScanSite 3 (http://scansite3.mit.edu). Site numbers refer to human LIN28A. (e) Western blot analysis of LIN28A (S200) phosphorylation in a panel of human PSCs. A representative image of two independent experiments is shown. Unprocessed scans of blots are shown in Supplementary Fig. 5.

Supplementary Figure 2 LIN28A and let-7 levels after MEK/ERK inhibitor treatment.

(a) Western blot (left) and qRT-PCR (right) analysis of transgenic FLAG-LIN28A in HeLa-LIN28A cells after 48-hour treatment with DMSO or PD0325901 (1 μM). n = 3 independent experiments. Error bars represent s.e.m. n.s. = non-significant; P = 0.19 (two-tailed Student’s t-test). (b) qRT-PCR analysis of pri/pre- (left) and mature (right) let-7s in PA1 cells treated with DMSO or PD0325901 (1 μM) for 48 h. n = 3 independent experiments. Error bars represent s.e.m. P > 0.05 (two-tailed Student’s t-test, PD versus DMSO). Statistics source data are shown in Supplementary Table 5. Unprocessed scans of blots are shown in Supplementary Fig. 5.

Supplementary Figure 3 Effects of mild LIN28A depletion on let-7 and LIN28’s direct mRNA targets.

qRT-PCR analysis of mature let-7s (a) and LIN28A’s mRNA targets (b), and Western blot analysis of LIN28A’s mRNA targets (c) in PA1 cells after mild LIN28A knockdown. siNC = negative control siRNA. n = 3 independent experiments. Error bars represent s.e.m. P > 0.05 (two-tailed Student’s t-test, siLIN28A versus siNC). Statistics source data are shown in Supplementary Table 4. Unprocessed scans of blots are shown in Supplementary Fig. 5.

Supplementary Figure 4 Effects of LIN28A level and S200 phosphorylation on let-7 and LIN28A’s direct mRNA targets.

(a) Corresponding levels of mature let-7s (top) and transgenic FLAG-LIN28A protein (middle and bottom) in a clonal series of HeLa-LIN28A cells. PA1 cells are included as a reference for native levels of LIN28A in hPSCs. Data are representative of two independent experiments. (b) Western blot analysis of transgenic FLAG-LIN28A in HeLa Flp-In cells stably expressing Dox-inducible wild-type (WT) or phospho-null (S200A) FLAG-LIN28A, without or with treatment with Dox. Dox = doxycycline (100 ng ml−1). A representative image of three independent experiments is shown. (c) qRT-PCR analysis of mature let-7 species in HeLa Flp-In cells stably expressing Dox-inducible wild-type (WT) or phospho-null (S200A) LIN28A, without or with treatment with Dox. Dox = doxycycline (100 ng ml−1).n = 3 independent experiments. Error bars represent s.e.m. P > 0.05 (two-tailed Student’s t-test). (d) Quantification of immunoprecipitated RNA (>200 nt) from HeLa Flp-In cells stably expressing wild-type (WT) or phospho-null (S200A) LIN28A. Western blot validation of the immunoprecipitation is shown on the bottom. n = 3 independent experiments. Error bars represent s.e.m. P < 0.05 (two-tailed Student’s t-test). (e) RNA-seq analysis of mRNAs immunoprecipitated by wild-type (WT) or phospho-null (S200A) FLAG-LIN28A in HeLa Flp-In cells. Each dot represents an average enrichment value for transcripts from a given gene. n = 3 independent experiments. Data were normalized to the amount of immunoprecipitated LIN28A prior to sequencing. Detailed description of the analysis is provided in the Methods section and the complete data set is available in Supplementary Table 3. (f) Western blot analysis of RNA immunoprecipitation in HeLa Flp-In cells overexpressing wild-type (WT) or phospho-mimetic (S200E) FLAG-LIN28A. Cells were treated with doxycycline (100 ng ml−1) to induce FLAG-LIN28A 48 h prior to analysis. A representative image of three independent experiments is shown. (g) qRT-PCR analysis of representative mRNA targets immunoprecipitated by wild-type (WT) or phospho-mimetic (S200E) FLAG-LIN28A in HeLa Flp-In cells. n = 3 independent experiments. Data were normalized to cell number prior to RT. Error bars represent s.e.m. P < 0.05; P < 0.01 (two-tailed Student’s t-test, S200E versus WT). Statistics source data are shown in Supplementary Table 4. Unprocessed scans of blots are shown in Supplementary Fig. 5.

Supplementary information

Supplementary Information

Supplementary Information (PDF 3658 kb)

Supplementary Table 1

Supplementary Information (XLSX 9 kb)

Supplementary Table 2

Supplementary Information (XLSX 3395 kb)

Supplementary Table 3

Supplementary Information (XLSX 3246 kb)

Supplementary Table 4

Supplementary Information (XLSX 59 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tsanov, K., Pearson, D., Wu, Z. et al. LIN28 phosphorylation by MAPK/ERK couples signalling to the post-transcriptional control of pluripotency. Nat Cell Biol 19, 60–67 (2017). https://doi.org/10.1038/ncb3453

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3453

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