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:

Sequential regulatory loops as key gatekeepers for neuronal reprogramming in human cells

Abstract

Direct conversion of somatic cells into neurons holds great promise for regenerative medicine. However, neuronal conversion is relatively inefficient in human cells compared to mouse cells. It has been unclear what might be the key barriers to reprogramming in human cells. We recently elucidated an RNA program mediated by the polypyrimidine tract binding protein PTB to convert mouse embryonic fibroblasts (MEFs) into functional neurons. In human adult fibroblasts (HAFs), however, we unexpectedly found that invoking the documented PTB–REST–miR-124 loop generates only immature neurons. We now report that the functionality requires sequential inactivation of PTB and the PTB paralog nPTB in HAFs. Inactivation of nPTB triggers another self-enforcing loop essential for neuronal maturation, which comprises nPTB, the transcription factor BRN2, and miR-9. These findings suggest that two separate gatekeepers control neuronal conversion and maturation and consecutively overcoming these gatekeepers enables deterministic reprogramming of HAFs into functional neurons.

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: Quantitative neuronal induction by shPTB in combination with small molecule inhibitors on HAFs.
Figure 2: Generation of functional neurons from HAFs by sequential knockdown of PTB and nPTB.
Figure 3: Neuronal maturation induced by BRN2 in combination with PTB knockdown.
Figure 4: BRN2 is essential to neuronal maturation in hNPCs and activation of mature-neuron-specific miRNAs.
Figure 5: Specific miRNAs repress nPTB during neuronal differentiation.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Hobert, O. Regulation of terminal differentiation programs in the nervous system. Annu. Rev. Cell Dev. Biol. 27, 681–696 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Molyneaux, B.J., Arlotta, P., Menezes, J.R. & Macklis, J.D. Neuronal subtype specification in the cerebral cortex. Nat. Rev. Neurosci. 8, 427–437 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Guillemot, F. Spatial and temporal specification of neural fates by transcription factor codes. Development 134, 3771–3780 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Bang, A.G. & Goulding, M.D. Regulation of vertebrate neural cell fate by transcription factors. Curr. Opin. Neurobiol. 6, 25–32 (1996).

    Article  CAS  PubMed  Google Scholar 

  5. Florio, M. et al. Human-specific gene ARHGAP11B promotes basal progenitor amplification and neocortex expansion. Science 347, 1465–1470 (2015).

    Article  CAS  PubMed  Google Scholar 

  6. Herculano-Houzel, S. The human brain in numbers: a linearly scaled-up primate brain. Front. Hum. Neurosci. 3, 31 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Lepski, G. et al. Delayed functional maturation of human neuronal progenitor cells in vitro. Mol. Cell. Neurosci. 47, 36–44 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  9. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. Dimos, J.T. et al. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science 321, 1218–1221 (2008).

    Article  CAS  PubMed  Google Scholar 

  12. Wernig, M. et al. Neurons derived from reprogrammed fibroblasts functionally integrate into the fetal brain and improve symptoms of rats with Parkinson’s disease. Proc. Natl. Acad. Sci. USA 105, 5856–5861 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Caiazzo, M. et al. Direct generation of functional dopaminergic neurons from mouse and human fibroblasts. Nature 476, 224–227 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Pang, Z.P. et al. Induction of human neuronal cells by defined transcription factors. Nature 476, 220–223 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Pfisterer, U. et al. Direct conversion of human fibroblasts to dopaminergic neurons. Proc. Natl. Acad. Sci. USA 108, 10343–10348 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Son, E.Y. et al. Conversion of mouse and human fibroblasts into functional spinal motor neurons. Cell Stem Cell 9, 205–218 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Vierbuchen, T. et al. Direct conversion of fibroblasts to functional neurons by defined factors. Nature 463, 1035–1041 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Broccoli, V., Caiazzo, M., & Dell'Anno, M.T. Setting a highway for converting skin into neurons. J. Mol. Cell Biol. 3, 322–323 (2011).

    Article  PubMed  Google Scholar 

  19. Amamoto, R. & Arlotta, P. Development-inspired reprogramming of the mammalian central nervous system. Science 343, 1239882 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Cahan, P. et al. CellNet: network biology applied to stem cell engineering. Cell 158, 903–915 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Morris, S.A. et al. Dissecting engineered cell types and enhancing cell fate conversion via CellNet. Cell 158, 889–902 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hou, P. et al. Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science 341, 651–654 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Li, Y. et al. Generation of iPSCs from mouse fibroblasts with a single gene, Oct4, and small molecules. Cell Res. 21, 196–204 (2011).

    Article  CAS  PubMed  Google Scholar 

  24. Cimadamore, F., Amador-Arjona, A., Chen, C., Huang, C.T. & Terskikh, A.V. SOX2-LIN28/let-7 pathway regulates proliferation and neurogenesis in neural precursors. Proc. Natl. Acad. Sci. USA 110, E3017–E3026 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Li, X. & Jin, P. Roles of small regulatory RNAs in determining neuronal identity. Nat. Rev. Neurosci. 11, 329–338 (2010).

    Article  CAS  PubMed  Google Scholar 

  26. Ambasudhan, R. et al. Direct reprogramming of adult human fibroblasts to functional neurons under defined conditions. Cell Stem Cell 9, 113–118 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yoo, A.S. et al. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature 476, 228–231 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Xue, Y. et al. Direct conversion of fibroblasts to neurons by reprogramming PTB-regulated microRNA circuits. Cell 152, 82–96 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Chambers, S.M. et al. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat. Biotechnol. 27, 275–280 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Li, W. et al. Rapid induction and long-term self-renewal of primitive neural precursors from human embryonic stem cells by small molecule inhibitors. Proc. Natl. Acad. Sci. USA 108, 8299–8304 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Liu, M.L. et al. Small molecules enable neurogenin 2 to efficiently convert human fibroblasts into cholinergic neurons. Nat. Commun. 4, 2183 (2013).

    Article  PubMed  CAS  Google Scholar 

  32. Ladewig, J. et al. Small molecules enable highly efficient neuronal conversion of human fibroblasts. Nat. Methods 9, 575–578 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Prasad, K.N. & Hsie, A.W. Morphologic differentiation of mouse neuroblastoma cells induced in vitro by dibutyryl adenosine 3′:5′-cyclic monophosphate. Nat. New Biol. 233, 141–142 (1971).

    Article  CAS  PubMed  Google Scholar 

  34. Boutz, P.L. et al. A post-transcriptional regulatory switch in polypyrimidine tract-binding proteins reprograms alternative splicing in developing neurons. Genes Dev. 21, 1636–1652 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zheng, S. et al. PSD-95 is post-transcriptionally repressed during early neural development by PTBP1 and PTBP2. Nat. Neurosci. 15, 381–388 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Li, Q. et al. The splicing regulator PTBP2 controls a program of embryonic splicing required for neuronal maturation. Elife 3, e01201 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Hagino-Yamagishi, K. et al. Predominant expression of Brn-2 in the postmitotic neurons of the developing mouse neocortex. Brain Res. 752, 261–268 (1997).

    Article  CAS  PubMed  Google Scholar 

  38. Marchetto, M.C. et al. A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell 143, 527–539 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dominguez, M.H., Ayoub, A.E. & Rakic, P. POU-III transcription factors (Brn1, Brn2, and Oct6) influence neurogenesis, molecular identity, and migratory destination of upper-layer cells of the cerebral cortex. Cereb. Cortex 23, 2632–2643 (2013).

    Article  PubMed  Google Scholar 

  40. Shibata, M., Nakao, H., Kiyonari, H., Abe, T. & Aizawa, S. MicroRNA-9 regulates neurogenesis in mouse telencephalon by targeting multiple transcription factors. J. Neurosci. 31, 3407–3422 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Krichevsky, A.M., King, K.S., Donahue, C.P., Khrapko, K. & Kosik, K.S. A microRNA array reveals extensive regulation of microRNAs during brain development. RNA 9, 1274–1281 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Makeyev, E.V., Zhang, J., Carrasco, M.A. & Maniatis, T. The MicroRNA miR-124 promotes neuronal differentiation by triggering brain-specific alternative pre-mRNA splicing. Mol. Cell 27, 435–448 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Åkerblom, M. et al. MicroRNA-124 is a subventricular zone neuronal fate determinant. J. Neurosci. 32, 8879–8889 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Giusti, S.A. et al. MicroRNA-9 controls dendritic development by targeting REST. Elife 3 (2014).

  45. Shibasaki, T. et al. PTB deficiency causes the loss of adherens junctions in the dorsal telencephalon and leads to lethal hydrocephalus. Cereb. Cortex 23, 1824–1835 (2013).

    Article  PubMed  Google Scholar 

  46. Gao, Z. et al. The master negative regulator REST/NRSF controls adult neurogenesis by restraining the neurogenic program in quiescent stem cells. J. Neurosci. 31, 9772–9786 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Spellman, R., Llorian, M. & Smith, C.W. Crossregulation and functional redundancy between the splicing regulator PTB and its paralogs nPTB and ROD1. Mol. Cell 27, 420–434 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Licatalosi, D.D. et al. Ptbp2 represses adult-specific splicing to regulate the generation of neuronal precursors in the embryonic brain. Genes Dev. 26, 1626–1642 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Rakic, P. Evolution of the neocortex: a perspective from developmental biology. Nat. Rev. Neurosci. 10, 724–735 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Xue, Y. et al. Genome-wide analysis of PTB-RNA interactions reveals a strategy used by the general splicing repressor to modulate exon inclusion or skipping. Mol. Cell 36, 996–1006 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sánchez, S. et al. A cAMP-activated pathway, including PKA and PI3K, regulates neuronal differentiation. Neurochem. Int. 44, 231–242 (2004).

    Article  PubMed  CAS  Google Scholar 

  52. Langmead, B., & Salzberg, S.L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank Z.-N. Zhang from A.R. Muotri's laboratory (University of California, San Diego) for sharing hNPCs and culture protocols and K. Zhang (University of California, San Diego) for sharing various antibodies and GFP transgenic rats. We are grateful to S. Dowdy for help on FACS analysis, to J. Ravits (University of California, San Diego) for human adult fibroblasts, and to Y. Yu from G. Hannon's laboratory (Cold Spring Harbor Laboratory) for the pTRIPZ lentiviral plasmid. This work was supported by a grant from the National Natural Science Foundation of China (91440101) to Y.X., by key programs of Chinese Academy of Sciences to X.-D.F. and Y.X. (KJZD-EW-L12) and by NIH grants (GM049369 and HG004659) to X.-D.F.

Author information

Authors and Affiliations

Authors

Contributions

Y.X. and X.-D.F. conceived the project. Y.X. designed and performed most biochemical experiments. H.Q. was responsible for all electrophysiology analysis and hNPC differentiation with input from A.K. and Z.P. B.Z. performed bioinformatics analysis and Y.Z. and X.H. contributed to RNA-seq analysis. J.H. performed EdU labeling, ChIP-qPCR, and FACS analysis. Y.X. and X.-D.F. wrote the manuscript.

Corresponding authors

Correspondence to Yuanchao Xue or Xiang-Dong Fu.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 PTB knockdown in human adult fibroblasts dampens cell proliferation and cell fate switch to the neuronal lineage.

(a) Characterization of HAFs by immunostaining with various cell type markers. HAFs show homogeneous staining of the fibroblast markers Fibronectin, FSP1 and Vimentin, but not markers for keratinocytes (K5, P63), melanocytes (MITF, P75), neural crest derivatives (SOX10, BRN3A) and neurons (indicated in the lower panel). n=2 biological repeats.

(b) FAC analysis of EdU-pulse labeled HAFs with or without PTB knockdown. Nuclei were stained by DAPI and EdU positive cells were stained with Alexa fluor 647. n=3 biological repeats.

(c) Example of Tuj1 (Green) positive cells that lack EdU (Red) staining. n=3 biological repeats.

(d) Example of induced neuronal-like cells by PTB knockdown only or in combination with specific small molecules. Red: Tuj1 expression, Blue: DAPI-stained nuclei. Scale bar: 20 µm. n=2 biological repeats.

(e) Phase contrast pictures of control and PTB shRNA-treated HAFs with small molecules treatment at different time points. The 3 small molecules were either present through the assay periods or washed away from the first treatment for 3 hrs. n=2 biological repeats.

(f) Immunostaining of the neural crest markers Nestin and Sox2 during PTB knockdown induced neuronal conversion. Scale bar: 100 µm. n=3 biological repeats.

Supplementary Figure 2 Validation of nPTB knockdown efficiency by RT-qPCR and Western blotting.

(a) Immunostaining of NeuN and NF (neurofilament) expression after different days of PTB knockdown. Scale bar: 50 µm. n=3 independent experiments.

(b) Relative expression of PTB and nPTB examined by RNA-seq analysis during neuronal conversion from HAFs. n=2 biological repeats.

(c, d) The mRNA and protein levels of nPTB assessed on shPTB-treated HAFs by RT-qPCR after the addition of doxycycline for 4 days to induce shnPTB expression. Beta-Actin (ACTB) served as loading control. Data are presented as mean± SD. Two-tailed unpaired Student’s t test was applied to calculate significance. n=3 independent experiments. NS: υ=4, t=0.628, P=0.564; 1#: υ=4, t=16.752, P=0.0000744; 2#: υ=4, t=5.872, P= 0.00420); 3#: υ=4, t=27.311, P= 0.0000107; 4#: υ=4, t=39.992, P=0.00000234. **P<0.01, ***P<0.001. Uncropped versions of Western blots are shown in Supplementary Figure 7.

Supplementary Figure 3 ChIP-qPCR analysis of REST targets and characterization of calcium influx in response to membrane depolarization in induced neurons with shPTB plus various neuronal lineage-specific transcription factors.

(a) ChIP-qPCR analysis of REST binding at the promoters of indicated neuronal transcription factors and miRNA loci in HAFs. Two-tailed unpaired Student’s t-test was applied to calculate significance. Data are presented as mean± SD. n=3 independent experiments. Ascl1: υ=4, t=5.336, P=0.00594; Myt1l: υ=4, t=11.605, P=0.000315); NeuroD1: υ=4, t=34.602, P=0.000004; Olig2: υ=4, t=12.332, P=0.000248; Brn2: υ=4, t=3.737, P=0.020; miR-124: υ=4, t= 5.812, P=0.00436. *P<0.05, **P<0.01, ***P<0.001.

(b) Screening for transcription factors capable of promoting MAP2 expression (Green) when combined with PTB knockdown in HAFs.

(c) Calcium influx in neuronal cells induced by co-expression of shPTB and BRN2 in HAFs. Scale bar: 30 µm.

Supplementary Figure 4 BRN2 regulation of a diverse repertoire of targets to control neuronal maturation.

(a) Characterization of hNPCs by immunostaining Nestin (Green), BRN2 (Red) and SOX2 (Red).

(b) Immunofluorescence analysis of neurons derived from hNPCs after BRN2 knockdown.

(c) The impact of BRN2 knockdown on the expression of MAP2 (Green) and SOX2 (Red) in neurons derived from hNPCs. n= 3 independent experiments for (a), (b) and (c). Scale bar: 20 µm.

(d) Immunostaining of NeuN after knockdown of BRN2 in PTB/nPTB sequentially depleted HAFs. n=3 random 20x fields.

(e) Impaired Na+ (Red) and K+ (Blue) currents in BRN2 knockdown neurons from hNPCs.

(f) Western blotting analysis of MAP2, Tuj1 and BRN2 in differentiated hNPCs. GAPDH served as loading control. n= 2 biological repeats. Uncropped versions of Western blots are shown in Supplementary Figure 7.

(g) Test of 4 commercially available anti-BRN2 antibodies by immunoprecipitation. C-20 and B-2 were chosen for ChIP-seq library construction. GAPDH served as input control. Uncropped versions of Western blots are shown in Supplementary Figure 7.

(h, i) GO term analysis of BRN2 target genes. Both GO terms of biological processes (g) and biological components (h) indicate BRN2 is critical for neuronal maturation. Background was based on all expressed genes in hNPCs.

Supplementary Figure 5 Regulation of nPTB by BRN2.

(a) Western blot analysis of PTB and nPTB expression in hNPC derived neurons upon depletion of BRN2. Beta-Actin and GAPDH served as loading controls. n=3 independent experiments. Two-tailed unpaired Student’s t test was applied to calculate significance. Data are presented as mean± SD. υ=4, t=13.208, P=0.000190. ***P<0.001.

(b) Western blot analysis of nPTB expression upon the ectopic expression of BRN2. Beta-Actin served as loading control. n=3 independent experiments. Uncropped versions of Western blots are shown in Supplementary Figure 7.

Supplementary Figure 6 Two consecutive RNA loops to control neuronal conversion and maturation in HAFs.

The previously deduced PTB-miR-124-REST loop for neuronal conversion28 and the newly elucidated nPTB-miR-9-BRN2 loop for neuronal maturation. Each component of the loops likely regulates a set of downstream targets to generate specific neuronal phenotypes.

Supplementary Figure 7 Uncropped versions of western blots.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Table 1 (PDF 2148 kb)

Supplementary Methods Checklist

(PDF 453 kb)

Supplementary Table 2

BRN2 ChIP-seq targets in the human genome (XLSX 257 kb)

Supplementary Table 3

PCR primers used in this study (XLSX 14 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xue, Y., Qian, H., Hu, J. et al. Sequential regulatory loops as key gatekeepers for neuronal reprogramming in human cells. Nat Neurosci 19, 807–815 (2016). https://doi.org/10.1038/nn.4297

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4297

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