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:

Reprogramming roadmap reveals route to human induced trophoblast stem cells

Abstract

The reprogramming of human somatic cells to primed or naive induced pluripotent stem cells recapitulates the stages of early embryonic development1,2,3,4,5,6. The molecular mechanism that underpins these reprogramming processes remains largely unexplored, which impedes our understanding and limits rational improvements to reprogramming protocols. Here, to address these issues, we reconstruct molecular reprogramming trajectories of human dermal fibroblasts using single-cell transcriptomics. This revealed that reprogramming into primed and naive pluripotency follows diverging and distinct trajectories. Moreover, genome-wide analyses of accessible chromatin showed key changes in the regulatory elements of core pluripotency genes, and orchestrated global changes in chromatin accessibility over time. Integrated analysis of these datasets revealed a role for transcription factors associated with the trophectoderm lineage, and the existence of a subpopulation of cells that enter a trophectoderm-like state during reprogramming. Furthermore, this trophectoderm-like state could be captured, which enabled the derivation of induced trophoblast stem cells. Induced trophoblast stem cells are molecularly and functionally similar to trophoblast stem cells derived from human blastocysts or first-trimester placentas7. Our results provide a high-resolution roadmap for the transcription-factor-mediated reprogramming of human somatic cells, indicate a role for the trophectoderm-lineage-specific regulatory program during this process, and facilitate the direct reprogramming of somatic cells into induced trophoblast stem 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

Fig. 1: Charting a roadmap for reprogramming human cells.
Fig. 2: Distinct transcriptional regulatory programs drive primed and naive human reprogramming.
Fig. 3: Derivation of iTS cells during reprogramming.
Fig. 4: Direct derivation of iTS cells from human fibroblasts.

Similar content being viewed by others

Data availability

We developed an interactive online tool (http://hrpi.ddnetbio.com/) to facilitate exploration of the dataset, and for downloading all of the processed datasets. Raw and processed next-generation sequencing datasets have been deposited at the NCBI Gene Expression Omnibus (GEO) repository under accession numbers: GSE150311 (scRNA-seq experiments of intermediates during human primed and naive reprogramming); GSE150637 (scRNA-seq experiments of day 21 reprogramming intermediates cultured under fibroblast condition, naive pluripotent and trophoblast stem cell conditions); GSE147564 (snRNA-seq experiments of intermediates during human primed and naive reprogramming); GSE147641 (ATAC-seq experiments of intermediates during human primed and naive reprogramming); GSE150590 (ATAC-seq experiments of iTS cells); GSE149694 (bulk RNA-seq experiments of intermediates during human primed and naive reprogramming); and GSE150616 (bulk RNA-seq experiments of iTS cells and their derived placenta subtypes). Source data are provided with this paper.

Code availability

All data were analysed with standard programs and packages as detailed. Scripts can be found at https://github.com/SGDDNB/hrpi.

References

  1. Gafni, O. et al. Derivation of novel human ground state naive pluripotent stem cells. Nature 504, 282–286 (2013).

    Article  ADS  CAS  PubMed  Google Scholar 

  2. Theunissen, T. W. et al. Systematic identification of culture conditions for induction and maintenance of naive human pluripotency. Cell Stem Cell 15, 524–526 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Takashima, Y. et al. Resetting transcription factor control circuitry toward ground-state pluripotency in human. Cell 162, 452–453 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Liu, X. et al. Comprehensive characterization of distinct states of human naive pluripotency generated by reprogramming. Nat. Methods 14, 1055–1062 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Kilens, S. et al. Parallel derivation of isogenic human primed and naive induced pluripotent stem cells. Nat. Commun. 9, 360 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  6. Giulitti, S. et al. Direct generation of human naive induced pluripotent stem cells from somatic cells in microfluidics. Nat. Cell Biol. 21, 275–286 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Okae, H. et al. Derivation of human trophoblast stem cells. Cell Stem Cell 22, 50–63.e6 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. O’Malley, J. et al. High-resolution analysis with novel cell-surface markers identifies routes to iPS cells. Nature 499, 88–91 (2013).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  10. Chronis, C. et al. Cooperative binding of transcription factors orchestrates reprogramming. Cell 168, 442–459.e20 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Knaupp, A. S. et al. Transient and permanent reconfiguration of chromatin and transcription factor occupancy drive reprogramming. Cell Stem Cell 21, 834–845.e6 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Schiebinger, G. et al. Optimal-transport analysis of single-cell gene expression identifies developmental trajectories in reprogramming. Cell 176, 1517 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Takahashi, K. et al. Induction of pluripotency in human somatic cells via a transient state resembling primitive streak-like mesendoderm. Nat. Commun. 5, 3678 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  14. Cacchiarelli, D. et al. Integrative analyses of human reprogramming reveal dynamic nature of induced pluripotency. Cell 162, 412–424 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Wang, Y. et al. Unique molecular events during reprogramming of human somatic cells to induced pluripotent stem cells (iPSCs) at naïve state. eLife 7, e29518 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Jacomy, M., Venturini, T., Heymann, S. & Bastian, M. ForceAtlas2, a continuous graph layout algorithm for handy network visualization designed for the Gephi software. PLoS ONE 9, e98679 (2014).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  17. Haghverdi, L., Buettner, F. & Theis, F. J. Diffusion maps for high-dimensional single-cell analysis of differentiation data. Bioinformatics 31, 2989–2998 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Gulati, G. S. et al. Single-cell transcriptional diversity is a hallmark of developmental potential. Science 367, 405–411 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  19. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wolf, F. A. et al. PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells. Genome Biol. 20, 59 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  22. O’Brien, C. M. et al. New monoclonal antibodies to defined cell surface proteins on human pluripotent stem cells. Stem Cells 35, 626–640 (2017).

    Article  PubMed  Google Scholar 

  23. Kumar, L. & E Futschik, M. Mfuzz: a software package for soft clustering of microarray data. Bioinformation 2, 5–7 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Yan, L. et al. Single-cell RNA-seq profiling of human preimplantation embryos and embryonic stem cells. Nat. Struct. Mol. Biol. 20, 1131–1139 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. Petropoulos, S. et al. Single-cell RNA-seq reveals lineage and X chromosome dynamics in human preimplantation embryos. Cell 165, 1012–1026 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lee, C. Q. E. et al. What is trophoblast? A combination of criteria define human first-trimester trophoblast. Stem Cell Reports 6, 257–272 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Vento-Tormo, R. et al. Single-cell reconstruction of the early maternal–fetal interface in humans. Nature 563, 347–353 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Turco, M. Y. et al. Trophoblast organoids as a model for maternal–fetal interactions during human placentation. Nature 564, 263–267 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Haider, S. et al. Self-renewing trophoblast organoids recapitulate the developmental program of the early human placenta. Stem Cell Reports 11, 537–551 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Dong, C. et al. Derivation of trophoblast stem cells from naïve human pluripotent stem cells. eLife 9, e52504 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Soufi, A. et al. Pioneer transcription factors target partial DNA motifs on nucleosomes to initiate reprogramming. Cell 161, 555–568 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Gao, X. et al. Establishment of porcine and human expanded potential stem cells. Nat. Cell Biol. 21, 687–699 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Posfai, E., Schell, J. P., Janiszewski, A., Rovic, I. & Murray, A. Defining totipotency using criteria of increasing stringency. Preprint at https://www.biorxiv.org/content/10.1101/202003.02.972893v1 (2020).

  34. Liu, X., Nefzger, C. & Polo, J. Establishment and maintenance of human naive pluripotent stem cells by primed to naive conversion and reprogramming of fibroblasts. Protoc. Exch. https://doi.org/10.1038/protex.2017.099 (2017).

  35. Guo, G. et al. Naive pluripotent stem cells derived directly from isolated cells of the human inner cell mass. Stem Cell Reports 6, 437–446 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Pastor, W. A. et al. Naive human pluripotent cells feature a methylation landscape devoid of blastocyst or germline memory. Cell Stem Cell 18, 323–329 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Larcombe, M. R. et al. Production of high-titer lentiviral particles for stable genetic modification of mammalian cells. Methods Mol. Biol. 1940, 47–61 (2019).

    Article  CAS  PubMed  Google Scholar 

  38. Qiu, P. et al. Extracting a cellular hierarchy from high-dimensional cytometry data with SPADE. Nat. Biotechnol. 29, 886–891 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nefzger, C. M. et al. A versatile strategy for isolating a highly enriched population of intestinal stem cells. Stem Cell Reports 6, 321–329 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Meistermann, D. et al. Spatio-temporal analysis of human preimplantation development reveals dynamics of epiblast and trophectoderm. Preprint at https://www.biorxiv.org/content/10.1101/604751v1 (2019).

  41. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Yang, S. et al. Decontamination of ambient RNA in single-cell RNA-seq with DecontX. Genom. Biol. 21, 57 (2020).

    Article  Google Scholar 

  43. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  48. Grubman, A., Choo, X. Y., Chew, G., Ouyang, J. F. & Sun, G. Mouse and human microglial phenotypes in Alzheimer’s disease are controlled by amyloid plaque phagocytosis through Hif1α. Preprint at https://www.biorxiv.org/content/10.1101/639054v1 (2019).

  49. Girardot, C., Scholtalbers, J., Sauer, S., Su, S.-Y. & Furlong, E. E. M. Je, a versatile suite to handle multiplexed NGS libraries with unique molecular identifiers. BMC Bioinformatics 17, 419 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  50. 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  CAS  PubMed  PubMed Central  Google Scholar 

  51. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 10 (2011).

    Article  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Lindenbaum, P. JVarkit: java-based utilities for Bioinformatics, https://figshare.com/articles/JVarkit_java_based_utilities_for_Bioinformatics/1425030 (2015).

  54. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  ADS  Google Scholar 

  55. Feng, J., Liu, T., Qin, B., Zhang, Y. & Liu, X. S. Identifying ChIP-seq enrichment using MACS. Nat. Protoc. 7, 1728–1740 (2012).

    Article  CAS  PubMed  Google Scholar 

  56. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Alasoo, K. et al. Transcriptional profiling of macrophages derived from monocytes and iPS cells identifies a conserved response to LPS and novel alternative transcription. Sci. Rep. 5, 12524 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  58. Wu, J. et al. Chromatin analysis in human early development reveals epigenetic transition during ZGA. Nature 557, 256–260 (2018).

    Article  ADS  CAS  PubMed  Google Scholar 

  59. Pastor, W. A. et al. TFAP2C regulates transcription in human naive pluripotency by opening enhancers. Nat. Cell Biol. 20, 553–564 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    ADS  PubMed  PubMed Central  Google Scholar 

  61. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Cavalcante, R. G. & Sartor, M. A. annotatr: genomic regions in context. Bioinformatics 33, 2381–2383 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank staff at Monash Flowcore Facility for providing high-quality cell sorting services and technical input; S. Wang, T. Wilson and the University of Melbourne Centre for Cancer Research (UMCCR) core for assistance with next-generation library preparation and Illumina sequencing; J. Hatwell-Humble for assistance with the mouse work; and A. Purcell for providing the HLA antibodies. We acknowledge the use of the services and facilities of Micromon, Monash Micro Imaging and Monash Histology Platforms at Monash University. This work was supported by National Health and Medical Research Council (NHMRC) project grants APP1104560 to J. M. Polo and A. L. Laslett, APP1069830 to R.L., and a Monash University strategic grant awarded to C.M.N. X.L. was supported by the Monash International Postgraduate Research Scholarship, a Monash Graduate Scholarship and the Carmela and Carmelo Ridolfo Prize in Stem Cell Research. A.S.K. was supported by an NHMRC Early Career Fellowship APP1092280. J. M. Polo and R.L. were supported by Silvia and Charles Viertel Senior Medical Research Fellowships. J. M. Polo was also supported by an ARC Future Fellowship FT180100674. R.L was supported by a Howard Hughes Medical Institute International Research Scholarship. O.J.L.R. and J.F.O. were supported by a Singapore National Research Foundation Competitive Research Programme (NRF-CRP20-2017-0002). The Australian Regenerative Medicine Institute is supported by grants from the State Government of Victoria and the Australian Government.

Author information

Authors and Affiliations

Authors

Contributions

J. M. Polo conceptualised the study. O.J.L.R. and J. M. Polo supervised the study. X.L., J.F.O., F.J.R., O.J.L.R. and J. M. Polo designed the experiments and analysis. O.J.L.R devised the single-cell analysis pipeline and data integration. X.L. performed reprogramming experiments, collection and isolation of single cells, intermediates and functional validation of iTS cell experiments with support from C.M.N., J.P.T., K.C.D., D.S.V., Y.B.Y.S., J.C., J. M. Paynter, J.F., Z.H., P.T., P.P.D. and S.K.N.; X.L. and C.M.N. performed single-cell RNA-seq, FACS experiments, SPADE analysis and the molecular experiments of the cells with support from A.S.K. and J.C.; L.G.M. helped with snRNA-seq experiments with support from A. L. Leichter; M.R.L. helped with RT-PCR experiments. D.P. helped with sequencing of day-21 reprogramming intermediates scRNA-seq libraries. X.L. generated the lentiviral particles with the assistance of J.P.T., G.S.; J.P. helped with ATAC-seq experiments. H.S.C., C.M.O’B. and A. L. Laslett. provided reagents and technical assistance. H.N. and D.R.P helped with bulk RNA-seq analysis. J.F.O. performed the snRNA-seq, scRNA-seq and bulk RNA-seq analyses for the human reprogramming intermediates and iTS cell experiments as well as the integration across the various datasets with support from F.J.R., J.S., J. M. Polo and O.J.L.R.; F.J.R. performed ATAC-seq analysis with support from V.T., X.Y.C, J.S., S.B., O.J.L.R., W.A.P., D.C., A.T.C., J. M. Polo and R.L.; J.F.O. and O.J.L.R. developed the interface for the interactive online tool. X.L., J.F.O., F.J.R., O.J.L.R. and J. M. Polo wrote the manuscript with input from K.C.D., A.G., A.T.C., L.D., C.M.N. and R.L. All authors approved of and contributed to, the final version of the manuscript.

Corresponding authors

Correspondence to Owen J. L. Rackham or Jose M. Polo.

Ethics declarations

Competing interests

O.J.L.R. and J. M. Polo. are co-inventors on a patent (WO/2017/106932) and are co-founders and shareholders of Mogrify Ltd., a cell therapy company. X.L., J.F.O., K.C.D., L.D., O.J.L.R. and J. M. Polo are co-inventors on a provisional patent application (application number: 2019904283) filed by Monash University, National University of Singapore and Université de Nantes related to work on derivation of iTS cells. The other authors declare no competing interests.

Additional information

Peer review information Nature thanks Ashley Moffett, Samantha A. Morris and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Fig. 1 Experimental designs, analysis pipelines for snRNA-seq and scRNA-seq.

a, Morphological changes of cells undergoing reprogramming in fibroblast medium (FM); primed reprogramming (PR); naive reprogramming (NR). FM: D0, 3 and 7; PR: D13, D21 and iPS cells (iPSCs); and NR: D13, D21 and iPS cells, n > 10. Scale bar, 500 μm. b, Immunostaining at early stages (FM: D0, 3 and 7), during PR (D13 and D21) and NR (D13 and D21) with TRA-1-60 for primed colonies, KLF17 for naive colonies and DAPI for nuclei staining, n = 2. Scale bar, 50 μm. c, Experimental design for scRNA-seq libraries. iMEF, irradiated mouse embryonic fibroblasts. d, snRNA-seq and scRNA-seq data analysis strategy (Methods). e, Representation of integrated snRNA-seq and scRNA-seq experiments (43,791 cells) on FDL. f, g, Primed and naive libraries on FDL. h, FDL showing cells in predicted stages of the cell cycle. i, Reprogramming trajectories on FDL highlighting cells within each time point. j, Expression of genes associated with primed pluripotency (NLGN4X) and naive pluripotency (DPPA5) on FDL. kr, PCA (kp), diffusion maps (q) and UMAP (r) of snRNA-seq and scRNA-seq data. For more details on sample numbers and statistics, see ‘Statistics and reproducibility’ in Methods.

Source data

Extended Data Fig. 2 Resolving the molecular hallmarks of primed and naive reprogramming trajectories.

a, Unsupervised clustering projected onto the FDL shown in Fig. 1 (43,791 cells). fm1–fm6, fibroblast and early reprogramming intermediate cell clusters; mix, shared cell cluster; pr1–pr3, primed reprogramming cell clusters; nr1–nr4, naive reprogramming cell clusters; nic, novel intermediate cell cluster; re1–re6, refractory cell clusters. b, snRNA-seq time point and library contribution (composition and cell number) towards each cell cluster. c, PAGA trajectory inference on diffusion maps. d, snRNA-seq clusters, used to define gene signatures, on FDL. e, Dot plot showing the expression of mesenchymal and epithelial (MET)-associated genes across cell clusters. f, Jaccard similarity of snRNA-seq cluster-specific genes. Cluster-specific genes are then grouped to define the eight gene signatures, highlighted at the bottom. g, Defined gene signatures on FDL. h, Gene-expression heat map of the primed or naive pluripotency signatures across the cell clusters (coloured arrows indicate known marker genes). i, Area plots showing the transition and activation of the defined signatures during primed and naive reprogramming over time. For more details on sample numbers and statistics, see ‘Statistics and reproducibility’ in Methods.

Source data

Extended Data Fig. 3 Isolation and characterization of intermediates during reprogramming into several naive human induced pluripotent states.

a, Identification of cell-surface markers for the isolation of primed and naive reprogramming intermediates. b, PCA of bulk RNA-seq data of isolated intermediates during primed and naive reprogramming, n ≥ 2. c, Experimental designs for the generation, isolation and profiling of intermediates during reprogramming into several naive human induced pluripotent states. d, Morphological changes during reprogramming under naive 5iLAF, NHSM and RSeT culture conditions (Methods), n = 4. Scale bar, 500 μm. e, Visualization of flow cytometry profiles (SPADE tree) of intermediates during reprogramming, n = 2. f, PCA of RNA-seq of primed and several types of naive reprogramming intermediates (Methods), n ≥ 2. g, Heat map showing gene expression profiles of primed and naive pluripotency signatures genes (defined in snRNA-seq and scRNA-seq analysis) across reprogramming intermediates and iPS cells derived under all different culture conditions, n ≥ 2. For more details on sample numbers and statistics, see ‘Statistics and reproducibility’ in Methods.

Source data

Extended Data Fig. 4 Single-cell profiling of the reprogramming pathway into naive RSeT state.

a, FDL of fibroblast, primed, naive t2iLGoY and RSeT scRNA-seq libraries (9,852 cells) (Methods). b, Expression profile of genes associated with human fibroblasts (ANPEP), shared pluripotency (NANOG), primed pluripotency (ZIC2 and NLGN4X) and naive pluripotency (DNMT3L and DPPA5) on FDL. For more details on sample numbers and statistics, see ‘Statistics and reproducibility’ in Methods.

Source data

Extended Data Fig. 5 Dynamics of chromatin state transitions during reprogramming into primed and naive human induced pluripotency.

a, PCA plot of ATAC-seq nucleosome-free signals, PC1 versus PC3 related to Fig. 2c. ATAC-seq was performed using isolated reprogramming intermediates and iPS cells from FM (D0, D3 and D7), PR (D13, D21, P3 and P10), NR (D13, D21, P3 and P10), n = 2. FM, fibroblasts medium (black); PR, primed reprogramming (orange); NR, naive reprogramming (blue). b, c, PCA plot of the integration of RNA-seq and ATAC-seq experiments (n ≥ 2). d, e, ATAC-seq and corresponding RNA-seq tracks of primed and naive reprogramming intermediates for fibroblast marker, ANPEP; shared pluripotency marker, PRDM14; primed-specific pluripotency marker SOX11; naive-specific pluripotency marker DNMT3L. Model of each gene is shown: coding sequences, light blue boxes, and exons, dark blue boxes; introns are shown as light blue connecting lines. f, Naive-reprogramming-specific ATAC-seq signals (in light grey) around core pluripotency factors NANOG and POU5F1 (also known as OCT4), naive-reprogramming-specific KLF17 and ZNF729 in primed and naive reprogramming intermediates and iPS cells compared to human inner cell mass and primed ES cells (ESCs) ATAC-seq data58. For more details on sample numbers, see ‘Statistics and reproducibility’ in Methods.

Source data

Extended Data Fig. 6 Features of accessible chromatin landscape during reprogramming into primed and naive human induced pluripotency.

a, Proportion of genomic regions in each of the ATAC-seq clusters. b, Averaged chromatin accessibility (z-scaled, n = 2) and gene expression (z-scaled, n ≥ 2) of one representative gene from each of the ATAC-seq peak clusters. c, Standardized gene expression (averaged z-scaling) of genes associated with ATAC-seq cluster peaks (Methods). d, Transcription factor motif enrichment analysis of the ATAC-seq peak clusters. Motif enrichment (−log(P value)) heat map by colour and the size the percentage of sequences in the cluster featuring the motif. Red arrow points to OCT4, SOX2, NANOG and KLF4 motifs in transient ATAC-seq cluster (C3), Blue arrow = enrichment of TE-associated transcription factors TFAP2C and GATA2 (C7 and C8) are indicated by blue arrows. e, Gene-expression heat map transcription factors identified in the motif enrichment analysis in d. f, TFAP2C and GATA2 gene expression during primed and naive reprogramming. g, Reverse transcription qPCR analysis of shTFAP2C and shGATA2 compared to scrambled controls, n = 2. For more details on sample numbers and statistics, see ‘Statistics and reproducibility’ in Methods.

Source data

Extended Data Fig. 7 Uncovering the transcriptional programmes of human fibroblast reprogramming into naive induced pluripotency.

a, b, Primed and naive scores, using gene signatures defined in this study (Fig. 1g), on human preimplantation embryos at indicated embryonic stages based on scRNA-seq experiments from published studies24,25. c, EPI, PE and TE signatures score at indicated embryonic stages25. d, EPI, PE and TE gene signatures25 from embryonic (E) day 5, 6 and 7 on intermediates and iPS cells reprogrammed under primed and different naive culture conditions (Methods). e, Gene set enrichment analysis (GSEA) (Methods) of the EPI, PE and TE gene signatures in reprogramming intermediates and iPS cells reprogrammed under primed and several naive culture conditions. f, EPI, PE and TE gene signatures scores in reprogramming intermediates and iPS cells reprogrammed under primed and several naive culture conditions. We used a combined gene signature across E5 to E7 for each lineage (Methods). g, EPI and PE signatures on FDL with single-cell trajectories constructed using Monocle3 (43,791 cells), related to Fig. 3a. h, Scoring of novel-intermediate signatures defined in this study (Extended Data Fig. 2f, g) on human preimplantation embryos of different lineages at indicated embryonic stages based on scRNA-seq experiments from published studies24,25. For more details on sample numbers and statistics, see ‘Statistics and reproducibility’ in Methods.

Source data

Extended Data Fig. 8 Characterization of iTSCd21n.

a, Immunostaining of fibroblast, primed, naive t2iLGoY iPS cells with P63, TFAP2C, GATA2 and KRT7, n = 2. Scale bar, 100 μm. b, Gene expression of trophoblast genes in fibroblasts, primed, naive t2iLGoY iPS cells, iTSCd21n and TS cells (TSCs) derived from a human blastocyst (TSCblast)7 and first-trimester placental trophoblast (TSCCT)7, mean of replicates, n = 2. c, Phase-contrast image of ST and EVT cells differentiated from iTSCd21n, n = 4. Scale bar, 100 μm. d, Fusion index of iTSCd21n ST and iTSCd21n, n = 5, data are represented as mean ± s.e.m. P values by two-tailed unpaired Student’s t-test. e, Representative results for over-the-counter hCG pregnancy test for medium of ST cells differentiated from iTSCd21n and control medium, n = 6. f, hCG levels in iTSCd21n- and iTSCd21n-ST conditioned medium, detected by ELISA, n = 4. g, hCG level in mouse blood serum detected by ELISA, n = 4. h, Lesions collected from subcutaneously engrafted iTSCd21n in NOD-SCID mice, n = 4. i, Haematoxylin and eosin, and immunohistochemical staining of KRT7 in the lesions from h. No evident lesions were observed in vehicle controls, n = 4. Scale bar, 200 μm. j, Distinct level of CD70 expression in naive and TE populations (indicated by blue arrows) on FDL projection of snRNA-seq and scRNA-seq datasets. k, Quantification of KRT7+ colony clusters after 9 d of transitioning into TS cell medium of unenriched, CD70high and CD70low populations, n = 2 or 3 independent experiments, data are mean ± s.e.m. P values by two-tailed unpaired Student’s t-test. Representative images of whole-well scans (top panels) (scale bar, 1 mm) and KRT7 immunostaining (bottom panels) (scale bar, 100 μm). For more details on sample numbers and statistics, see ‘Statistics and reproducibility’ in Methods.

Source data

Extended Data Fig. 9 Cellular heterogeneity of fibroblast and iTSCd8 reprogramming intermediates revealed by scRNA-seq.

a, Experimental designs and preparation of scRNA-seq libraries of day-21 fibroblast, naive and TSCd8 reprogramming intermediates. b, Strength of EPI signatures on FDL (10,518 cells). The cell population not enriched for EPI signatures but enriched for TE signatures is indicated by a purple arrow, related to Fig. 4b. c, Representation of 13 cell clusters from unsupervised clustering projected onto the FDL, fibroblast medium cell clusters: D21fm1–D21fm7; naive reprogramming cell clusters: D21nr1–D21nr3; trophoblast reprogramming cell clusters: D21tr1–D21tr3. d, Contribution of each scRNA-seq library (%) to the composition of cell clusters. D21tr1 cluster is indicated by a purple arrow. e, Expression of genes associated with human fibroblasts (ANPEP), shared pluripotency (NANOG), primed pluripotency (ZIC2), naive pluripotency (DNMT3L) and trophoblast (GATA3) on FDL projection of day-21 fibroblast, naive and TSCd8 reprogramming intermediate scRNA-seq libraries (top panels). Defined fibroblast, early-primed, primed, novel-intermediate and naive signatures (Extended Data Fig. 2f) on the FDL projection (bottom panels). f, Experimental designs to validate the potential of day-21 fibroblast reprogramming intermediates for the derivation of primed, naive iPS cells and iTS cells. g, Phase-contrast images of primed, naive iPS cells and iTS cells generated from day 21 fibroblast reprogramming intermediates, n = 2. Scale bar, 50 μm. Immunostaining of primed, naive iPS cells and iTS cells with NANOG, KLF17, NR2F2, KRT7 and DAPI for nuclei staining, n = 2. Scale bar, 200 μm. h, Reverse-transcription qPCR analysis of NANOG, ZIC2, KLF17, DPPA3, GATA2 and KRT7 expression in primed, naive iPS cells and iTS cells generated from day-21 fibroblast reprogramming intermediates, n = 3. Data are mean ± s.e.m. For more details on sample numbers and statistics, see ‘Statistics and reproducibility’ in Methods.

Source data

Extended Data Fig. 10 Characterization of iTSCd8.

a, Sendai viral transgenes in iTS cell lines with positive and negative controls, n = 6. b, Gene expression of trophoblast genes in fibroblasts, primed iPS cells, naive t2iLGoY iPS cells, iTSCd8 and iTSCd21n compared to TSCs derived from a human blastocyst (TSCblast) and first-trimester placental trophoblast (TSCCT)7, data are presented as mean (n = 2). c, Cell fusion index of iTSCd8 ST and iTSCd8. n = 5, data are mean ± s.e.m. P values by two-tailed unpaired Student’s t-test. d, Representative results for hCG pregnancy test obtained from medium of ST cells differentiated from iTSCd8, n = 6. e, hCG levels of iTSCd8- and iTSCd8-ST-conditioned medium detected by ELISA, n = 4. f, Representative flow cytometry analysis of pan HLA-A, B, C class I marker (W6/32), HLA-Bw4 and HLA-G in fibroblasts and EVTs, n = 4. g, Representative flow cytometry analysis of pan HLA class I marker (W6/32) and HLA-G in iTSCd8 EVT and iTSCd21n EVT. h, Representative flow cytometry analysis of pan HLA class I marker (W6/32) in fibroblasts, primed iPS cells, naive t2iLGoY iPS cells, iTSCd8 and iTSCd21n, n = 4. i, j, Expression of ST genes in iTSCd8- and iTSCd21n-derived ST cells (i) and expression of EVT genes in iTSCd8 and iTSCd21n-derived EVT cells (j). k, l, Spearman correlation of the transcriptomes of fibroblast, primed and naive t2iLGoY iPS cells, iTSCd8 and iTSCd21n, iTSCd8 ST and iTSCd21n ST, iTSCd8 EVT and iTSCd21n EVT generated in this study with trophoblast organoids samples from refs. 28,29 (k) and single-cell fetal–maternal interface samples from ref. 27 (l), n ≥ 2, replicates are averaged before performing correlation. m, Lesions collected from subcutaneously engrafted iTSCd8 in NOD-SCID mice, n = 4. For more details on sample numbers and statistics, see ‘Statistics and reproducibility’ in Methods.

Source data

Supplementary information

41586_2020_2734_MOESM1_ESM.pdf

Supplementary Figure 1 Isolation of reprogramming intermediates using a panel of cell surface markers. a, Flow cytometry analysis of TRA-1-60 vs SSEA3, EPCAM vs SSEA3 over the reprogramming time-course into primed and naive induced pluripotency. b, Validation of the panel of cell surface markers for isolation of intermediates that carry the reprogramming potential across the reprogramming time-course. e.g., CD13+F11R- and CD13+F11R+ subpopulations were isolated on day 3 and reseeded for 5 days (for flow cytometry reanalysis) and for hiPSCs colony formation (AP staining), see Methods for details. Of note, SSEA3+EPCAM+ population on day 13 of NR showed negative for AP staining due to the substantial differentiation of these cells after reseeding. FM: Fibroblasts Medium (black); PR: Primed Reprogramming (orange); NR: Naive Reprogramming (blue).

Reporting Summary

Supplementary Tables

This zipped file contains Supplementary Tables 1-16 and a guide to the tables.

41586_2020_2734_MOESM4_ESM.mp4

Video 1 Dynamics of cellular transitions during primed and naive human reprogramming. Video showing individual snRNA-seq libraries at each time during primed and naive reprogramming.

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, X., Ouyang, J.F., Rossello, F.J. et al. Reprogramming roadmap reveals route to human induced trophoblast stem cells. Nature 586, 101–107 (2020). https://doi.org/10.1038/s41586-020-2734-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2734-6

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