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:

NKX3-1 is required for induced pluripotent stem cell reprogramming and can replace OCT4 in mouse and human iPSC induction

Abstract

Reprogramming somatic cells to induced pluripotent stem cells (iPSCs) is now routinely accomplished by overexpression of the four Yamanaka factors (OCT4, SOX2, KLF4, MYC (or OSKM))1. These iPSCs can be derived from patients’ somatic cells and differentiated toward diverse fates, serving as a resource for basic and translational research. However, mechanistic insights into regulators and pathways that initiate the pluripotency network remain to be resolved. In particular, naturally occurring molecules that activate endogenous OCT4 and replace exogenous OCT4 in human iPSC reprogramming have yet to be found. Using a heterokaryon reprogramming system we identified NKX3-1 as an early and transiently expressed homeobox transcription factor. Following knockdown of NKX3-1, iPSC reprogramming is abrogated. NKX3-1 functions downstream of the IL-6–STAT3 regulatory network to activate endogenous OCT4. Importantly, NKX3-1 substitutes for exogenous OCT4 to reprogram both mouse and human fibroblasts at comparable efficiencies and generate fully pluripotent stem cells. Our findings establish an essential role for NKX3-1, a prostate-specific tumour suppressor, in iPSC reprogramming.

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

Fig. 1: Nuclear reprogramming of human fibroblasts after fusion with mouse ESCs.
Fig. 2: Motifs at early accessible chromatin and gene expression dynamics identify NKX3-1.
Fig. 3: NKX3-1 induces Oct4 expression and is required for iPSC formation.
Fig. 4: NKX3-1 can substitute for OCT4 to generate pluripotent mouse and human iPSCs.
Fig. 5: NKX3-1 functions downstream of the IL-6–STAT3 cascade during iPSC induction.

Similar content being viewed by others

References

  1. Takahashi, K. & Yamanaka, S. A decade of transcription factor-mediated reprogramming to pluripotency. Nat. Rev. Mol. Cell Biol. 17, 183–193 (2016).

    Article  PubMed  CAS  Google Scholar 

  2. Buganim, Y. et al. Single-cell expression analyses during cellular reprogramming reveal an early stochastic and a late hierarchic phase. Cell 150, 1209–1222 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Heng, J.-C. D. et al. The nuclear receptor Nr5a2 can replace Oct4 in the reprogramming of murine somatic cells to pluripotent cells. Cell Stem Cell 6, 167–174 (2010).

    Article  PubMed  CAS  Google Scholar 

  4. Eguchi, A. et al. Reprogramming cell fate with a genome-scale library of artificial transcription factors. Proc. Natl Acad. Sci. USA 113, E8257–E8266 (2016).

    Article  PubMed  CAS  Google Scholar 

  5. Gao, Y. et al. Replacement of Oct4 by Tet1 during iPSC induction reveals an important role of DNA methylation and hydroxymethylation in reprogramming. Cell Stem Cell 12, 453–469 (2013).

    Article  PubMed  CAS  Google Scholar 

  6. Long, Y., Wang, M., Gu, H. & Xie, X. Bromodeoxyuridine promotes full-chemical induction of mouse pluripotent stem cells. Cell Res. 25, 1171–1174 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  8. Redmer, T. et al. E-cadherin is crucial for embryonic stem cell pluripotency and can replace OCT4 during somatic cell reprogramming. EMBO Rep. 12, 720–726 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Tan, F., Qian, C., Tang, K., Abd-Allah, S. M. & Jing, N. Inhibition of transforming growth factor β (TGF-β)signaling can substitute for Oct4 protein in reprogramming and maintain pluripotency. J. Biol. Chem. 290, 4500–4511 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Anokye-Danso, F. et al. Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell 8, 376–388 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Miyoshi, N. et al. Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell 8, 633–638 (2011).

    Article  PubMed  CAS  Google Scholar 

  12. Shu, J. et al. Induction of pluripotency in mouse somatic cells with lineage specifiers. Cell 153, 963–975 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Montserrat, N. et al. Reprogramming of human fibroblasts to pluripotency with lineage specifiers. Cell Stem Cell 13, 341–350 (2013).

    Article  PubMed  CAS  Google Scholar 

  14. Bhutani, N. et al. Reprogramming towards pluripotency requires AID-dependent DNA demethylation. Nature 463, 1042–1047 (2009).

    Article  CAS  Google Scholar 

  15. Brady, J. J. et al. Early role for IL-6 signalling during generation of induced pluripotent stem cells revealed by heterokaryon RNA-Seq. Nat. Cell Biol. 15, 1244–1252 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Bhattacharya, B. et al. Gene expression in human embryonic stem cell lines: unique molecular signature. Blood 103, 2956–2964 (2003).

    Article  PubMed  CAS  Google Scholar 

  17. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Ernst, J. & Kellis, M. ChromHMM: automating chromatin-state discovery and characterization. Nat. Methods 9, 215–216 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Nordhoff, V. et al. Comparative analysis of human, bovine, and murine Oct-4 upstream promoter sequences. Mamm. Genome 12, 309–317 (2001).

    Article  PubMed  CAS  Google Scholar 

  20. Jerabek, S., Merino, F., Schöler, H. R. & Cojocaru, V. OCT4: dynamic DNA binding pioneers stem cell pluripotency. Biochim Biophys. Acta 1839, 138–154 (2013).

    Article  PubMed  CAS  Google Scholar 

  21. Xu, Y. et al. Transcriptional control of somatic cell reprogramming. Trends Cell Biol. 26, 272–288 (2016).

    Article  PubMed  CAS  Google Scholar 

  22. Do, D. V. et al. A genetic and developmental pathway from STAT3 to the OCT4-NANOG circuit is essential for maintenance of ICM lineages in vivo. Genes Dev. 27, 1378–1390 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Dutta, A. et al. Identification of an NKX3.1-G9a-UTY transcriptional regulatory network that controls prostate differentiation. Science 352, 1576–1580 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Bhatia-Gaur, R. et al. Roles for Nkx3.1 in prostate development and cancer. Genes Dev. 13, 966–977 (1999).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Wang, X. et al. A luminal epithelial stem cell that is a cell of origin for prostate cancer. Nature 461, 495–500 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Qin, J. et al. The PSA(−/lo) prostate cancer cell population harbors self-renewing long-term tumor-propagating cells that resist castration. Cell Stem Cell 10, 556–569 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. He, W. W. et al. A novel human prostate-specific, androgen-regulated homeobox gene (NKX3.1) that maps to 8p21, a region frequently deleted in prostate cancer. Genomics 43, 69–77 (1997).

    Article  PubMed  CAS  Google Scholar 

  28. Dai, H.-Q. et al. TET-mediated DNA demethylation controls gastrulation by regulating Lefty-Nodal signalling. Nature 538, 528–532 (2016).

    Article  PubMed  CAS  Google Scholar 

  29. Mosteiro, L. et al. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science 354, aaf4445 (2016).

    Article  PubMed  CAS  Google Scholar 

  30. Rose-John, S. IL-6 trans-signaling via the soluble IL-6 receptor: importance for the pro-inflammatory activities of IL-6. Int J. Biol. Sci. 8, 1237–1247 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Bowen, C. & Gelmann, E. P. NKX3.1 activates cellular response to DNA damage. Cancer Res. 70, 3089–3097 (2010).

    Article  PubMed  CAS  Google Scholar 

  32. Banito, A. et al. Senescence impairs successful reprogramming to pluripotent stem cells. Genes Dev. 23, 2134–2139 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Gong, L. et al. p53 isoform Δ133p53 promotes efficiency of induced pluripotent stem cells and ensures genomic integrity during reprogramming. Sci. Rep. 6, 37281 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Eide, T., Ramberg, H., Glackin, C., Tindall, D. & Taskén, K. A. TWIST1, a novel androgen-regulated gene, is a target for NKX3-1 in prostate cancer cells. Cancer Cell Int. 13, 4 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Li, R. et al. A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell 7, 51–63 (2010).

    Article  PubMed  CAS  Google Scholar 

  36. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 12, 323 (2011).

    Article  CAS  Google Scholar 

  37. Kundaje, A. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

The authors apologize to those investigators whose important work we were unable to cite or describe due to space constraints. The authors thank D. Burns for critical discussions of the manuscript and P. Chu (Comparative Medicine, Stanford) for technical assistance. The authors acknowledge the Stanford Shared FACS Facility (SSFF) and FACS Core Facility in the Stanford Lokey Stem Cell Research Building for technical support. This work was supported by F32 GM112425-02, T32 HD007249 to T.M., Bio-X Graduate Research Fellowships to J.J.B., a NSF Graduate Research Fellowship to G.M., a GSK Sir James Black Program for Drug Discovery Postdoctoral Fellowship to A.P., and the Baxter Foundation, California Institute for Regenerative Medicine (CIRM) grant RB1-01292 and US National Institutes of Health (NIH) grants U01 HL100397, R01 AG009521 and R01 AG020961 to H.M.B.

Author information

Authors and Affiliations

Authors

Contributions

The study was designed by T.M. and H.M.B. T.M. performed the majority of the experiments. G.M., J.J.B., A.P., H.Z. and V.S. also performed experiments. T.M. and G.M. performed data analysis. The manuscript was written by T.M., G.M. and H.M.B.

Corresponding author

Correspondence to Helen M. Blau.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Dynamic transcriptional kinetics during early heterokaryon reprogramming.

(a) Schematic diagram of the heterokaryon time course. Heterokaryons were generated by fusion of mouse ESCs and human MRC5 fibroblasts in a 3:1 ratio respectively. Cells were collected at the indicated time points for RNA-seq. Gene expression analysis was performed using only human transcripts. (a) FACS sorting gating strategy to isolate hetorkaryons. Hetorkaryons are detected as dsRed and GFP double positive cells and are sorted to >90% purity. (c) Line plots showing clusters of gene expression patterns during heterokaryon reprogramming. Individual gene expression pattern (gray), the median pattern (red), trajectory, and Gene Ontology/Gene Identity classes. Data represent 1 of 3 biological experiments with similar results.

Supplementary Figure 2 Differential gene expression during early heterokaryon reprogramming.

(a) Scatter plot of log(Fold Change) and mean expression of genes. Red dots signify a DE gene.

Supplementary Figure 3 Nkx3-1 knock-down does not affect MEF proliferation or survival.

(a) Line plot showing growth curves of MEFs transduced with OSKM and an shctrl or shNKX3-1 lentivirus (n = 3 biological replicates). Unpaired Student’s t-test was used and data represents mean ± s.d. (b) Histogram plot showing the percentage of live cells in MEFs transduced with OSKM and an shctrl or shNKX3-1 lentivirus. Live cells were determined as having no 7-AAD staining (n = 3 biological replicates). Unpaired Student’s t-test was used and data represents mean ± s.d. Statistical source data and exact P values for a and b can be found in Supplementary Table 5.

Supplementary Figure 4 NSK-derived iPSCs and OSK-derived have comparable levels of pluripotency gene expression.

(a) Histogram showing numbers of iPSC colonies number 21 days after lentivirus transduction of plasmids encoding NS, NK, and NKS. N = NKX3-1, S = SOX2, K = KLF4 (n = 2 biological replicates). (b) Human OCT4 and NKX3-1 expression in OSK-iPSCs and NSK-iPSCs. (n = 1 biological replicate) (c) Intracellular flow cytometry shows OCT4 protein expression in OSK-iPSCs and NSK-iPSCs. (Data represent 1 of 2 biological replicates with similar results). (d) Expression heatmap of pluripotency genes in MEFs, mouse NSK-iPSCs, mouse OSK-iPSCs and mouse ESCs. (e) Expression heatmap of pluripotency genes in human fibroblasts, human NSK-iPSCs, human OSK-iPSCs and human ESCs.

Supplementary Figure 5 IL6R and NKX3-1 expression kinetics during heterokaryon reprogramming.

(a) Line plot showing IL6R and NKX3-1 expression kinetics during heterokaryon reprogramming (n = 3 biological replicates). Data represents mean ± s.d. (b) Histogram plot showing expression of IL6R and NKX3-1 after transduction of an shIL6R lentivirus 2 h post-fusion (n = 3 biological replicates). Unpaired Student’s t-test was used and data represents mean ± s.d. (c) Histogram plot showing efficiency of shIL6R knock-down in mouse and human fibroblasts after infection with various targeted shRNAs. RT-qPCR was performed 5 days post transduction. (n = 3 technical replicates). (d) ATAC-seq tracks at the NKX3-1 locus during heterokaryon reprogramming. (Data represent 1 of 3 biological replicates with similar results). (e) Histogram plot showing expression of Nkx3-1 following transduction of OSKM alone and with Cre lentivirus (n = 3 biological replicates). Unpaired Student’s t-test was used and data represents mean ± s.d. (f) Histogram plot showing expression of NKX3-1 in human fibroblast after transduction of lentivirus encoding NKX3-1 (n = 3 biological replicates). Unpaired two-sided Student’s t-test was used. (g, h) Histogram plot showing expression of specific MET or EMT genes after transduction of NKX3-1 (n = 3 biological replicates). Unpaired Student’s t-test was used and data are shown as mean ± s.d. Statistical source data and exact P values for b, e, and f-h can be found in Supplementary Table 5.

Electronic supplementary material

Supplementary Information

Supplementary Figures 1–5 and Supplementary Table legends

Reporting Summary

Supplementary Table 1

Top 100 DE genes in each time point ordered by adjusted P-value

Supplementary Table 2

Human ESC signature gene expression in heterokaryon by 48h

Supplementary Table 3

Gene expression profile of IL-6 associated genes

Supplementary Table 4

Oligos

Supplementary Table 5

Statistics source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mai, T., Markov, G.J., Brady, J.J. et al. NKX3-1 is required for induced pluripotent stem cell reprogramming and can replace OCT4 in mouse and human iPSC induction. Nat Cell Biol 20, 900–908 (2018). https://doi.org/10.1038/s41556-018-0136-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0136-x

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer