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:

Lineage conversion induced by pluripotency factors involves transient passage through an iPSC stage

Abstract

Brief expression of pluripotency-associated factors such as Oct4, Klf4, Sox2 and c-Myc (OKSM), in combination with differentiation-inducing signals, has been reported to trigger transdifferentiation of fibroblasts into other cell types. Here we show that OKSM expression in mouse fibroblasts gives rise to both induced pluripotent stem cells (iPSCs) and induced neural stem cells (iNSCs) under conditions previously shown to induce only iNSCs. Fibroblast-derived iNSC colonies silenced retroviral transgenes and reactivated silenced X chromosomes, both hallmarks of pluripotent stem cells. Moreover, lineage tracing with an Oct4-CreER labeling system demonstrated that virtually all iNSC colonies originated from cells transiently expressing Oct4, whereas ablation of Oct4+ cells prevented iNSC formation. Lastly, an alternative transdifferentiation cocktail that lacks Oct4 and was reportedly unable to support induced pluripotency yielded iPSCs and iNSCs carrying the Oct4-CreER-derived lineage label. Together, these data suggest that iNSC generation from fibroblasts using OKSM and other pluripotency-related reprogramming factors requires passage through a transient iPSC state.

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: Generation of iPSCs and iNSCs from MEFs under neural culture conditions.
Figure 2: Oct4 lineage tracing and ablation during iNSC induction.
Figure 3: Silencing of retroviral transgenes and X chromosome reactivation during iNSC induction.
Figure 4: An alternative transdifferentiation cocktail generates iNSCs and iPSCs via an Oct4+ intermediate stage.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Vierbuchen, T. & Wernig, M. Molecular roadblocks for cellular reprogramming. Mol. Cell 47, 827–838 (2012).

    Article  CAS  Google Scholar 

  2. Efe, J.A. et al. Conversion of mouse fibroblasts into cardiomyocytes using a direct reprogramming strategy. Nat. Cell Biol. 13, 215–222 (2011).

    Article  CAS  Google Scholar 

  3. Han, D.W. et al. Direct reprogramming of fibroblasts into neural stem cells by defined factors. Cell Stem Cell 10, 465–472 (2012).

    Article  CAS  Google Scholar 

  4. Kim, J. et al. Direct reprogramming of mouse fibroblasts to neural progenitors. Proc. Natl. Acad. Sci. USA 108, 7838–7843 (2011).

    Article  CAS  Google Scholar 

  5. Li, K. et al. Small molecules facilitate the reprogramming of mouse fibroblasts into pancreatic lineages. Cell Stem Cell 14, 228–236 (2014).

    Article  CAS  Google Scholar 

  6. Margariti, A. et al. Direct reprogramming of fibroblasts into endothelial cells capable of angiogenesis and reendothelialization in tissue-engineered vessels. Proc. Natl. Acad. Sci. USA 109, 13793–13798 (2012).

    Article  CAS  Google Scholar 

  7. Thier, M. et al. Direct conversion of fibroblasts into stably expandable neural stem cells. Cell Stem Cell 10, 473–479 (2012).

    Article  CAS  Google Scholar 

  8. Zhu, S. et al. Mouse liver repopulation with hepatocytes generated from human fibroblasts. Nature 508, 93–97 (2014).

    Article  CAS  Google Scholar 

  9. Orkin, S.H. & Hochedlinger, K. Chromatin connections to pluripotency and cellular reprogramming. Cell 145, 835–850 (2011).

    Article  CAS  Google Scholar 

  10. Stadtfeld, M., Maherali, N., Borkent, M. & Hochedlinger, K. A reprogrammable mouse strain from gene-targeted embryonic stem cells. Nat. Methods 7, 53–55 (2010).

    Article  CAS  Google Scholar 

  11. Stadtfeld, M., Maherali, N., Breault, D.T. & Hochedlinger, K. Defining molecular cornerstones during fibroblast to iPS cell reprogramming in mouse. Cell Stem Cell 2, 230–240 (2008).

    Article  CAS  Google Scholar 

  12. DeVeale, B. et al. Oct4 is required E7.5 for proliferation in the primitive streak. PLoS Genet. 9, e1003957 (2013).

    Article  Google Scholar 

  13. Polo, J.M. et al. A molecular roadmap of reprogramming somatic cells into iPS cells. Cell 151, 1617–1632 (2012).

    Article  CAS  Google Scholar 

  14. Capela, A. & Temple, S. LeX/ssea-1 is expressed by adult mouse CNS stem cells, identifying them as nonependymal. Neuron 35, 865–875 (2002).

    Article  Google Scholar 

  15. Greder, L.V. et al. Analysis of endogenous Oct4 activation during induced pluripotent stem cell reprogramming using an inducible Oct4 lineage label. Stem Cells 30, 2596–2601 (2012).

    Article  Google Scholar 

  16. Esteban, M.A. et al. Vitamin C enhances the generation of mouse and human induced pluripotent stem cells. Cell Stem Cell 6, 71–79 (2010).

    Article  CAS  Google Scholar 

  17. Maherali, N. et al. Directly reprogrammed fibroblasts show global epigenetic remodeling and widespread tissue contribution. Cell Stem Cell 1, 55–70 (2007).

    Article  CAS  Google Scholar 

  18. Sommer, C.A. et al. Induced pluripotent stem cell generation using a single lentiviral stem cell cassette. Stem Cells 27, 543–549 (2009).

    Article  CAS  Google Scholar 

  19. Schwarz, B.A., Bar-Nur, O., Silva, J.C. & Hochedlinger, K. Nanog is dispensable for the generation of induced pluripotent stem cells. Curr. Biol. 24, 347–350 (2014).

    Article  CAS  Google Scholar 

  20. Bar-Nur, O. et al. Small molecules facilitate rapid and synchronous iPSC generation. Nat. Methods 11, 1170–1176 (2014).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  22. Lujan, E., Chanda, S., Ahlenius, H., Sudhof, T.C. & Wernig, M. Direct conversion of mouse fibroblasts to self-renewing, tripotent neural precursor cells. Proc. Natl. Acad. Sci. USA 109, 2527–2532 (2012).

    Article  CAS  Google Scholar 

  23. Weissbein, U., Ben-David, U. & Benvenisty, N. Virtual karyotyping reveals greater chromosomal stability in neural cells derived by transdifferentiation than those from stem cells. Cell Stem Cell 15, 687–691 (2014).

    Article  CAS  Google Scholar 

  24. Maza, I. et al. Transient acquisition of pluripotency during somatic cell transdifferentiation with iPSC reprogramming factors. Nat. Biotechnol. doi:10.1038/nbt.3270 (22 June 2015).

  25. Chen, J. et al. Rational optimization of reprogramming culture conditions for the generation of induced pluripotent stem cells with ultra-high efficiency and fast kinetics. Cell Res. 21, 884–894 (2011).

    Article  CAS  Google Scholar 

  26. Chambers, I. et al. Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell 113, 643–655 (2003).

    Article  CAS  Google Scholar 

  27. van Oosten, A.L., Costa, Y., Smith, A. & Silva, J.C. JAK/STAT3 signalling is sufficient and dominant over antagonistic cues for the establishment of naive pluripotency. Nat. Commun. 3, 817 (2012).

    Article  Google Scholar 

  28. 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 

  29. Eminli, S., Utikal, J., Arnold, K., Jaenisch, R. & Hochedlinger, K. Reprogramming of neural progenitor cells into induced pluripotent stem cells in the absence of exogenous Sox2 expression. Stem Cells 26, 2467–2474 (2008).

    Article  CAS  Google Scholar 

  30. Hadjantonakis, A.K., Gertsenstein, M., Ikawa, M., Okabe, M. & Nagy, A. Non-invasive sexing of preimplantation stage mammalian embryos. Nat. Genet. 19, 220–222 (1998).

    Article  CAS  Google Scholar 

  31. Wu, S., Wu, Y. & Capecchi, M.R. Motoneurons and oligodendrocytes are sequentially generated from neural stem cells but do not appear to share common lineage-restricted progenitors in vivo. Development 133, 581–590 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Hochedlinger laboratory for critical evaluation and discussion of this manuscript. We thank E. Apostolou and S. Cheloufi for helpful comments and N. Maherali for suggesting the idea of X chromosome reactivation. We are also grateful to B. Payer and J. Lee for providing tail-tip fibroblasts carrying an X-linked GFP reporter and to A. Brack for sharing Rosa26-lsl-DTA mice. We are grateful to L. Prickett, M. Weglarz and K. Folz-Donahue at the MGH/HSCI flow cytometry core for their constant assistance and support. O.B.-N. is supported by a Gruss-Lipper postdoctoral fellowship from the EGL foundation. B.A.S. has been supported by a T32 (5-T32-CA-9216-33) grant and through a Postdoctoral Fellowship Award by the MGH Executive Committee on Research Fund for Medical Discovery. J.B. and I.L. are supported by Ruth L. Kirschstein F32 Post-doctoral Fellowships from the US National Institutes of Health (NIH) (1F32HD078029-01A1 to J.B. and 1F32HD079225-01A1 to I.L.). Support to K.H. is from the NIH (R01HD058013) and Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

O.B.-N. and K.H. conceived the experiments, interpreted results and wrote the manuscript. O.B.-N. conducted all reprogramming experiments, performed statistical analysis, bioinformatics analysis of expression data and generated figures; C.V. assisted in most reprogramming experiments and interpretation of results; A.G.S. and G.M. generated the BKSM lentiviral cassette; J.B. generated chimeric animals by blastocyst injections; B.A.S. conducted reprogramming experiments from FACS-sorted intermediates; I.L. performed allele-specific real-time PCR for an X-linked marker gene; A.J.H. confirmed specificity of the Oct4-CreER lineage tracing system in vivo.

Corresponding author

Correspondence to Konrad Hochedlinger.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Characterization of OKSM-iNSCs

(A) Gene expression levels of Sox1 and Sox2 in the indicated cell types by microarray gene expression analysis. (B) Dendrogram cluster analysis of global gene expression of indicated samples by expression microarrays. (C) Scatter plot analysis shows linear regression coefficient (R2) of global gene expression by microarray analysis between MEFs and OKSM-iNSCs and brain-derived NSCs and OKSM-iNSCs, respectively. (D) Functional annotation analysis of upregulated genes (2-fold or more) in OKSM-iNSCs vs. MEFs by gene expression microarray. Top categories are shown together with the number of genes and Benjamini-Hochberg-corrected p-value. (E) Gene expression levels of Nanog in the indicated cell types by microarray analysis.

Supplementary Figure 2 Molecular characterization of iPSCs generated in NSC media and propagated in ESC media

(A) Graph showing quantification of Oct4+ and Nanog+ iPSC-like colonies generated in NSC media. For each replicate, 3x104 cells were used (n=3 independent replicates; error bars, s.e.m. *P<0.05). (B) Representative immunofluorescence images showing co-localization of Oct4+ and Nanog+ in colonies emerging in NSC media. (C) Morphology of NSC media-derived iPSCs (upper panel) and alkaline phosphatase (AP) staining for an expanded iPSC clone (lower panel). (D) Representative images of immunofluorescence staining for Nanog in NSC media-derived iPSC clones #1-3. (E) Expression of pluripotency-associated surface markers in NSC media-derived iPSCs by live-staining immunofluorescence analysis. (F) Flow cytometry analysis of surface markers characteristic of nascent iPSCs (loss of Thy1, gain of Epcam) in rep-MEFs 6 days after dox administration in conventional NSC media. (G) Gene expression levels of Epcam in the indicated cell types by microarray gene expression analysis. Note that Epcam is not expressed in brain-NSCs or OKSM-iNSCs.

Supplementary Figure 3 Activity of the Oct4-CreER; R26-lsl-EYFP lineage tracing system in brain-derived NSCs

(A) Flow cytometric analysis of Oct4-CreER; R26-lsl-EYFP NSC cultures derived from the brains of five independent E13.5 embryos. Tamoxifen was administered to pregnant mice at E8.5 and brain-derived NSCs were recovered at day E13.5. EYFP expression was examined at passage 3 of in vitro culture. OKSM-iNSC clone #2 served as a positive control for EYFP expression. The PE-Cy7 channel was used to control for autofluorescence. (B) Representative immunofluorescence images show that the Oct4-CreER; R26-lsl-EYFP brain-derived NSC lines #1-5 express the NSC markers Sox1 and Sox2. (C) Flow cytometry analysis for uninduced Oct4-CreER; R26-lsl-EYFP brain-derived NSCs to confirm faithful regulation of the Oct4-CreER allele. Brain-NSCs were isolated at E13.5 and 4-OHT was added after 2 passages before performing flow cytometry analysis for EYFP. Note that no labeled cells were detected with or without 4-OHT in brain-NSCs of this genotype, indicating specificity of the Oct4-CreER allele. The PE-Cy7 channel was used to control for autofluorescence.

Supplementary Figure 4 Specificity of the Oct4-GFP and Oct4-CreER; R26-lsl-EYFP reporters

(A) Flow cytometry analysis for EYFP expression at the indicated time points of OKSM induction in Oct4-CreER; R26-lsl-EYFP MEFs. Note that no aberrant EYFP expression is detected after 2 and 4 days of OKSM induction (OKSM ind.). The PE-Cy7 channel was used to control for autofluorescence. (B) Flow cytometry analysis for Oct4-GFP reporter expression at the indicated time points of OKSM induction shows absence of GFP signal. The PE-Cy7 channel was used to control for autofluorescence. Note that more cells are EYFP+ in panel A than are GFP+ in panel B after 11 days of dox treatment and 5 days of dox withdrawal (13.6% vs. 0.14%) because differentiated cells remain labeled with the Oct4-CreER; R26-lsl-EYFP system, whereas they downregulate the Oct4-GFP reporter.

Supplementary Figure 5 Molecular characterization of iPSCs generated from Oct4-CreER; R26-lsl-EYFP MEFs in NSC media and propagated in ESC media

(A) iPSC clones remain EYFP- in the absence of 4-OHT (left panels) while they become EYFP+ in the presence of 4-OHT (right panel). (B) Representative immunofluorescence stains for Nanog and Oct4 in NSC media-derived iPSC clones generated from Oct4-CreER; R26-lsl-EYFP MEFs. (C) Flow cytometry analysis for EYFP expression in MEFs derived from chimeric embryos generated from EYFP+ iPSCs clone #6 (generated in conventional NSC media). Shown are MEF preparations established from 2 different E13.5 embryos and analyzed at passage 2. Non-chimeric EYFP- MEFs served as a negative control. The PE-Cy7 channel was used to control for autofluorescence.

Supplementary Figure 6 Characterization of iNSC subclones

(A) Representative images of iNSC sucblones #2-6 at passage 20, derived by sorting single cells from EYFP+ iNSCs expanded from a single colony. (B) Flow cytometry analysis for EYFP expression of OKSM-iNSC subclones #2-6 at passage 12. Note that all subclones are at least 98% EYFP+ at passage 12. The PE-Cy7 channel was used to control for autofluorescence. (C) Immunofluorescence images show staining for Nestin and Sox1 in OKSM-iNSC sucblones #1-6 at passage 22.

Supplementary Figure 7 Oct4 lineage tracing and ablation systems to track the origin of iNSCs

(A) Representative images of OKSM-iNSC (EYFP+) colonies generated with modified NSC media after 4-5 days of doxycycline (dox) induction, followed by 17 days of dox-independent growth. Note that images depicting neurites were taken at higher magnification than those of the corresponding EYFP+ OKSM-iNSCs. (B) Schematic of lineage ablation approach using Oct4-CreER; R26-lsl-DTA MEFs to test whether all iNSCs pass through an Oct4+ stage. (C) 4-OHT treatment triggers death of Oct4+ cells as judged by alkaline phosphatase staining (AP) of dox-independent iPSC colonies in ESC media. The presence of residual AP+ colonies in 4-OHT treated Oct4-CreER; R26-lsl-DTA cells likely represents partially reprogrammed iPSCs or iPSC colonies that failed to recombine the R26-lsl-DTA allele. (D) Quantification of AP+, dox-independent iPSC formation shown in (C). For each replicate, 3x104 cells were used (n=3 independent replicates; error bars, s.e.m.).

Supplementary Figure 8 Retroviral tdTomato expression in MEFs cultured in NSC media

(A) Flow cytometry analysis of MEFs infected with the retroviral pMXs-tdTomato vector. Shown are bulk cultures expressing tdTomato (left panel), sorted tdTomato+ MEFs (middle panel) and uninfected MEFs (right panel). The PacB channel was used to control for autofluorescence. (B) Sorted and explanted tdTomato+ MEFs retain tdTomato expression after growth in NSC media for 1 week (left panel). Uninfected MEFs are shown on the right. The PE-Cy7 channel was used to control for autofluorescence.

Supplementary Figure 9 Expression of the X-linked CMV-GFP reporter and X-linked Uba1 gene in female TTFs and derivative iNSCs

(A) Flow cytometry analysis of OKSM-iNSC clones generated either from sorted XaXiGFP (top) or XaGFPXi tail tip fibroblasts (TTFs) (bottom). Note that some iNSC clones partially lose CMV-GFP reporter expression after prolonged culture, regardless of their origin from XaXiGFP or XaGFPXi TTFs. The PE-Cy7 channel was used to control for autofluorescence. (B) Sorted XaXiGFP TTFs do not reactivate the CMV-GFP reporter located on the silent X chromosome after 2 weeks of culture in NSC media. A positive control for GFP expression is shown on the right. The APC channel was used to control for autofluorescence. (C) Allele-specific quantitative PCR analysis of the X-linked gene Uba1. Primers distinguishing between the maternal mus musculus musculus (left) and the paternal mus musculus castaneous (right) alleles of the Uba1 gene were used. Sorted GFP+ and GFP- TTFs served as controls.

Supplementary Figure 10 Effect of small molecules on the generation of iNSCs

(A) Quantification of Sox1+ iNSC colonies generated in the presence of ascorbic acid (AA) or ascorbic acid and GSK3βi (“AGi”). For each replicate, 3x104 cells were used (n=3 independent replicates; error bars, s.e.m. *P<0.05). (B) Representative immunofluorescence images showing a Sox1+ iNSC colony generated with AGi.

Supplementary Figure 11 Cloning of the BKSM polycistronic cassette.

(A) A schematic representation of the cloning strategy to generate the BKSM lentiviral vector cassette. A DNA fragment consisting of the Oct4 ORF, followed by an F2A peptide and part of the Klf4 ORF, was excised from the STEMCCA (OKSM) vector using NotI and BstZ17I and replaced by a fragment corresponding to Brn4-F2A-Klf4, generated by overlapping PCR. (B) Representative immunofluorescence images for Oct4 expression in MEFs 3 days after transduction with the tetOP-OKSM (top) and tetOP-BKSM (bottom) cassettes. Note that Oct4+ nuclear staining is detected only in cells that were infected with the tetOP-OKSM cassette. (C) Representative immunofluorescence images for Brn4 in MEFs 3 days following transduction with the tetOP-BKSM (top) or tetOP-OKSM (bottom) cassettes. Note that Brn4+ nuclear staining is detected only in cells that were infected with the tetOP-BKSM cassette.

Supplementary Figure 12 Characterization of BKSM-iNSCs

(A) Morphology of a BKSM-iNSC colony generated from Sox2-GFP MEFs (upper panel) and Sox2-GFP reporter expression (lower panel). (B) Scatter plot display of global microarray data of indicated samples, showing low linear regression coefficient (R2) of global gene expression between MEFs and BKSM-iNSCs and high R2 between BKSM-iNSCs and brain-derived NSCs. (C) Functional annotation of upregulated genes (2-fold or more) in BKSM-iNSCs vs. MEFs based on microarray data; top categories are shown together with the number of genes and Benjamini-Hochberg-corrected P-values.

Supplementary Figure 13 Characterization of BKSM-iPSCs

(A) Representative images of iPSCs generated with the BKSM cassette from Sox2-GFP MEFs (BKSM-iPSCs). (B) Expression of key pluripotency genes in BKSM-iPSCs based on microarray analysis. (C) Scatter plot analysis showing linear regression coefficient (R2) of global gene expression between MEFs and BKSM-iPSCs, and mouse ESCs and BKSM-iPSCs. (D) Teratoma sections from a BKSM-iPSC clone showing cell types representative of the three germ layers. (E) Activation of Sox2-GFP expression in the brain of a neonatal chimera generated from BKSM-iPSCs (derived from Sox2-GFP MEFs) (left panel) relative to a non-chimeric control (right panel).

Supplementary Figure 14 Reprogramming of BKSM-iNSCs into iPSCs

(A) Experimental design to generate iPSCs using a secondary system of reprogrammable BKSM-iNSCs. (B) Morphology of iPSC colony generated from BKSM-iNSCs. (C) Alkaline phosphatase (AP) staining of iPSCs derived from BKSM-iNSCs.

Supplementary Figure 15 Oct4 lineage tracing during induced cardiomyocyte (iCM) or induced neuron (iN) generation from MEFs

(A) Quantification of EYFP+ and Cardiac Troponin (cTnT)+ induced cardiomyocytes derived from Oct4-CreER; R26-lsl-EYFP MEFs upon brief OKSM expression. For each replicate, 5x105 cells were used (n=3 biological replicates; error bars, s.e.m. ** P<0.005 n.s., not significant P=0.07). (B) Representative immunofluorescence images showing co-staining for EYFP and cTnT expression in induced cardiomyocytes. (C) Schematic of the lineage tracing approach to test if iN-like cells derived from Oct4-CreER; R26-lsl-EYFP MEFs upon transduction with tetOP-Ascl1, tetOP-Brn2 and tetOP-Myt1l vectors pass through an Oct4+ stage. (D) Lack of EYFP expression in non-transduced (upper panel) or transduced (middle panel) MEFs grown in iN transdifferentiation medium for 12 days. Tuj1+ iN-like cells were detected exclusively in cultures transduced with tetOP-Ascl1, tetOP-Brn2 and tetOP-Myt1l vectors. We calculated the frequency of iN formation by counting Tuj1+ cells per DAPI+ cells in 8 random fields, indicating a reprogramming efficiency of 9%. (13 iN cells/142 DAPI+ cells) (E) Flow cytometry analysis of bulk cultures undergoing transdifferentiation. Note the lack of EYFP expression in transduced MEFs relative to positive control (EYFP+ OKSM-iNSC clone #2). The PE-Cy7 channel was used to control for autofluorescence.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15 (PDF 1180 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bar-Nur, O., Verheul, C., Sommer, A. et al. Lineage conversion induced by pluripotency factors involves transient passage through an iPSC stage. Nat Biotechnol 33, 761–768 (2015). https://doi.org/10.1038/nbt.3247

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3247

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