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:

A fast chemical reprogramming system promotes cell identity transition through a diapause-like state

Abstract

Cellular reprogramming by only small molecules holds enormous potentials for regenerative medicine. However, chemical reprogramming remains a slow process and labour intensive, hindering its broad applications and the investigation of underlying molecular mechanisms. Here, through screening of over 21,000 conditions, we develop a fast chemical reprogramming (FCR) system, which significantly improves the kinetics of cell identity rewiring. We find that FCR rapidly goes through an interesting route for pluripotent reprogramming, uniquely transitioning through a developmentally diapause-like state. Furthermore, FCR critically enables comprehensive characterizations using multi-omics technologies, and has revealed unexpected important features including key regulatory factors and epigenetic dynamics. Particularly, activation of pluripotency-related endogenous retroviruses via inhibition of heterochromatin significantly enhances reprogramming. Our studies provide critical insights into how only environmental cues are sufficient to rapidly reinstate pluripotency in somatic cells, and make notable technical and conceptual advances for solving the puzzle of regeneration.

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: Large-scale small molecule screens for CR.
Fig. 2: Establish an FCR platform.
Fig. 3: FCR is transcriptionally dynamic and goes through a diapause-like state.
Fig. 4: scRNA-seq characterization and examination of diapause-like state in FCR.
Fig. 5: Genome-wide characterization of epigenetic dynamics during FCR.
Fig. 6: Epigenetic remodelling on iPS cell-related ERVs.

Similar content being viewed by others

Data availability

All sequencing data generated during this study are available from the Gene Expression Omnibus (GEO) under the accession ID GSE188834. It contains bulk RNA-seq data (GSE188830 and GSE220554), ATAC-seq data (GSE188824), CUT&Tag data (GSE188826), RRBS data (GSE188831) and scRNA-seq data (GSE218855). Previously published TFR RNA-seq datasets were downloaded from GEO: GSE102518 and GSE125644. The accession numbers of two CR RNA-seq datasets are GSE73631 and GSE110264. The accession number of the diapause ICM RNA-seq dataset is GSE143494. The TFR ATAC-seq dataset was downloaded from GEO: GSE101905. The mouse genome used in this study: mm10 (https://www.ncbi.nlm.nih.gov/assembly/GCF_000001635.20/) and GRCm39 (https://www.ncbi.nlm.nih.gov/assembly/GCF_000001635.27/). The screen data have been deposited on figshare (https://doi.org/10.6084/m9.figshare.21959552). Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

References

  1. Gurdon, J. B. Adult frogs derived from nuclei of single somatic cells. Dev. Biol. 4, 256 (1962).

    Article  CAS  PubMed  Google Scholar 

  2. 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 

  3. Graf, T. Historical origins of transdifferentiation and reprogramming. Cell Stem Cell 9, 504–516 (2011).

    Article  CAS  PubMed  Google Scholar 

  4. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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 

  7. Stadhouders, R. et al. Transcription factors orchestrate dynamic interplay between genome topology and gene regulation during cell reprogramming. Nat. Genet. 50, 238–249 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Schiebinger, G. et al. Optimal-transport analysis of single-cell gene expression identifies developmental trajectories in reprogramming. Cell 176, 928–943.e22 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Zviran, A. et al. Deterministic somatic cell reprogramming involves continuous transcriptional changes governed by myc and epigenetic-driven modules. Cell Stem Cell 24, 328–341.e9 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Huangfu, D. et al. Induction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2. Nat. Biotechnol. 26, 1269–1275 (2008).

    Article  CAS  PubMed  Google Scholar 

  11. Ichida, J. K. et al. A small-molecule inhibitor of tgf-β signaling replaces sox2 in reprogramming by inducing nanog. Cell Stem Cell 5, 491–503 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Maherali, N. & Hochedlinger, K. Tgfbeta signal inhibition cooperates in the induction of iPSCs and replaces Sox2 and cMyc. Curr. Biol. 19, 1718–1723 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhu, S. et al. Reprogramming of human primary somatic cells by OCT4 and chemical compounds. Cell Stem Cell 7, 651–655 (2010).

    Article  CAS  PubMed  Google Scholar 

  14. 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 

  15. Li, W., Li, K., Wei, W. & Ding, S. Chemical approaches to stem cell biology and therapeutics. Cell Stem Cell 13, 270–283 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Theunissen, T. W. & Jaenisch, R. Molecular control of induced pluripotency. Cell Stem Cell 14, 720–734 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Xie, X., Fu, Y. & Liu, J. Chemical reprogramming and transdifferentiation. Curr. Opin. Genet Dev. 46, 104–113 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. Ma, X., Kong, L. & Zhu, S. Reprogramming cell fates by small molecules. Protein Cell 8, 328–348 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hou, P. 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 

  20. Zhao, Y. et al. A XEN-like state bridges somatic cells to pluripotency during chemical reprogramming. Cell 163, 1678–1691 (2015).

    Article  CAS  PubMed  Google Scholar 

  21. Zhao, T. et al. Single-cell RNA-seq reveals dynamic early embryonic-like programs during chemical reprogramming. Cell Stem Cell 23, 31–45.e7 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Cao, S. et al. Chromatin accessibility dynamics during chemical induction of pluripotency. Cell Stem Cell 22, 529–542.e5 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Jin, Y. et al. Harnessing endogenous transcription factors directly by small molecules for chemically induced pluripotency inception. Proc. Natl Acad. Sci. USA 120, e2215155120 (2023).

    Article  CAS  PubMed  Google Scholar 

  24. Huangfu, D. et al. Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat. Biotechnol. 26, 795–797 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ebrahimi, A. et al. Bromodomain inhibition of the coactivators CBP/EP300 facilitate cellular reprogramming. Nat. Chem. Biol. 15, 519–528 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang, W. Y. et al. Inhibition of Syk promotes chemical reprogramming of fibroblasts via metabolic rewiring and H2S production. EMBO J. https://doi.org/10.15252/embj.2020106771 (2021).

  27. Guan, J. et al. Chemical reprogramming of human somatic cells to pluripotent stem cells. Nature 605, 325–331 (2022).

    Article  CAS  PubMed  Google Scholar 

  28. Wu, Y. et al. Autophagy and mTORC1 regulate the stochastic phase of somatic cell reprogramming. Nat. Cell Biol. 17, 715–725 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Ma, T. et al. Atg5-independent autophagy regulates mitochondrial clearance and is essential for iPSC reprogramming. Nat. Cell Biol. 17, 1379–1387 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Li, Y. Y. et al. R-loops coordinate with SOX2 in regulating reprogramming to pluripotency. Sci. Adv. https://doi.org/10.1126/sciadv.aba0777 (2020).

  31. Renfree, M. B. & Shaw, G. Diapause. Annu. Rev. Physiol. 62, 353–375 (2000).

    Article  CAS  PubMed  Google Scholar 

  32. Bulut-Karslioglu, A. et al. Inhibition of mTOR induces a paused pluripotent state. Nature 540, 119–123 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Scognamiglio, R. et al. Myc depletion induces a pluripotent dormant state mimicking diapause. Cell 164, 668–680 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Hussein, A. M. et al. Metabolic control over mTOR-dependent diapause-like state. Dev. Cell 52, 236–250.e7 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Dhimolea, E. et al. An embryonic diapause-like adaptation with suppressed myc activity enables tumor treatment persistence. Cancer Cell 39, 240–256.e11 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Rehman, S. K. et al. Colorectal cancer cells enter a diapause-like DTP state to survive chemotherapy. Cell 184, 226–242.e21 (2021).

    Article  CAS  PubMed  Google Scholar 

  37. Salic, A. & Mitchison, T. J. A chemical method for fast and sensitive detection of DNA synthesis in vivo. Proc. Natl Acad. Sci. USA 105, 2415–2420 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liu, J., Xu, Y., Stoleru, D. & Salic, A. Imaging protein synthesis in cells and tissues with an alkyne analog of puromycin. Proc. Natl Acad. Sci. USA 109, 413–418 (2012).

    Article  CAS  PubMed  Google Scholar 

  39. Qin, H. et al. Systematic identification of barriers to human iPSC Generation. Cell 158, 449–461 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Fonseca, B. D. et al. La-related Protein 1 (LARP1) Represses Terminal Oligopyrimidine (TOP) mRNA Translation Downstream of mTOR Complex 1 (mTORC1). J. Biol. Chem. 290, 15996–16020 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Onder, T. T. et al. Chromatin-modifying enzymes as modulators of reprogramming. Nature 483, 598–602 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Li, D. et al. Chromatin accessibility dynamics during iPSC reprogramming. Cell Stem Cell 21, 819–833.e6 (2017).

    Article  CAS  PubMed  Google Scholar 

  43. Kiviet, D. J. et al. Stochasticity of metabolism and growth at the single-cell level. Nature 514, 376–379 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Matsui, T. et al. Proviral silencing in embryonic stem cells requires the histone methyltransferase ESET. Nature 464, 927–931 (2010).

    Article  CAS  PubMed  Google Scholar 

  45. Bulut-Karslioglu, A. et al. Suv39h-dependent H3K9me3 marks intact retrotransposons and silences LINE elements in mouse embryonic stem cells. Mol. Cell 55, 277–290 (2014).

    Article  CAS  PubMed  Google Scholar 

  46. Wang, C. et al. Reprogramming of H3K9me3-dependent heterochromatin during mammalian embryo development. Nat. Cell Biol. 20, 620–631 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Sridharan, R. et al. Proteomic and genomic approaches reveal critical functions of H3K9 methylation and heterochromatin protein-1γ in reprogramming to pluripotency. Nat. Cell Biol. 15, 872–882 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lu, Y. et al. Reprogramming to recover youthful epigenetic information and restore vision. Nature 588, 124–129 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Chen, Y. et al. Reversible reprogramming of cardiomyocytes to a fetal state drives heart regeneration in mice. Science 373, 1537–1540 (2021).

    Article  CAS  PubMed  Google Scholar 

  50. Browder, K. C. et al. In vivo partial reprogramming alters age-associated molecular changes during physiological aging in mice. Nat. Aging https://doi.org/10.1038/s43587-022-00183-2 (2022).

    Article  PubMed  Google Scholar 

  51. Zhao, X. Y. et al. iPS cells produce viable mice through tetraploid complementation. Nature 461, 86–90 (2009).

    Article  CAS  PubMed  Google Scholar 

  52. Legault, L. M., Chan, D. & McGraw, S. Rapid multiplexed reduced representation bisulfite sequencing library Prep (rRRBS). Bio Protoc. 9, e3171 (2019).

    PubMed  PubMed Central  Google Scholar 

  53. Kaya-Okur, H. S. et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat. Commun. 10, 1930 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Satpathy, A. T. et al. Massively parallel single-cell chromatin landscapes of human immune cell development and intratumoral T cell exhaustion. Nat. Biotechnol. 37, 925–936 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Van de Sande, B. et al. A scalable SCENIC workflow for single-cell gene regulatory network analysis. Nat. Protoc. 15, 2247–2276 (2020).

    Article  PubMed  Google Scholar 

  59. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Yu, G., Wang, L. G. & He, Q. Y. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, 2382–2383 (2015).

    Article  CAS  PubMed  Google Scholar 

  61. Krzywinski, M. et al. Circos: an information aesthetic for comparative genomics. Genome Res. 19, 1639–1645 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Ross-Innes, C. S. et al. Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 481, 389–393 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Akalin, A. et al. methylKit: a comprehensive R package for the analysis of genome-wide DNA methylation profiles. Genome Biol. https://doi.org/10.1186/gb-2012-13-10-R87 (2012).

  64. Jin, Y., Tam, O. H., Paniagua, E. & Hammell, M. TEtranscripts: a package for including transposable elements in differential expression analysis of RNA-seq datasets. Bioinformatics 31, 3593–3599 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of Zhu lab for helpful discussions. This work was supported by the grants from National Natural Science Foundation of China (nos. 32270781 and 31970818), the National Key Research and Development Program of China (no. 2016YFC1305300) and the Outstanding Youth Fund of Zhejiang Province (no. R17C120002).

Author information

Authors and Affiliations

Authors

Contributions

S.Z. and X.C. conceived and designed the project. X.C. performed the experiments. Y. Lu, X.C. and L.L. analysed the sequencing data. L.W. and W.L. performed the germline transmission experiment. X.M., J.P., Q.D., Y. Li, W.W., Y.J., Z.H., Z.Z., G.C., L.J., H.W., X.Z. and J.F. provided reagents and helped with experiments. X.H. and H.A.R. helped with discussing and editing the paper. S.Z., X.C. and Y. Lu wrote the paper with comments and contributions from all authors.

Corresponding author

Correspondence to Saiyong Zhu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks the anonymous reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Concentration tests.

af, Concentration test results of each small molecule and growth factor in D0-2 (a), D2-4 (b), D4-6 (c), D6-8 (d), D8-10 (e), D10-12 (f). Data are mean ± SD, n = 3 biological replicates.

Source data

Extended Data Fig. 2 Characterization of FCR-generated iPSCs.

a, Histogram showing the targets of small molecules. b, iPSC colony number of samples that medium was fused. Data are mean ± SD, n = 3 biological replicates. **P < 0.01; ***P < 0.001 (two-tailed Student’s t test). P values were 0.0026, 0.0025, 0.00042 from left to right. c, Expression of Oct4, Sox2, Rex1 and Nanog. Data are mean ± SD, n = 3 biological replicates. d, Volcano plots of differentially expressed genes. Red, up-regulated genes in iPSC; blue, down-regulated genes in iPSC; R, Pearson correlation coefficient. e, DNA methylation pattern in promotor regions of Oct4 and Nanog. f, Karyotype analysis of iPSC (passage 8). g, k, o, r, Representative fields from the indicated cells. Scale bars, 200 μm. h, l, p, s, Representative fields from FCR day 12 of the indicated cells. Scale bars, 200 μm. i, m, Expression of Oct4. Scale bars, 200 μm. j, n, q, t, Morphologies and pluripotency gene expression of iPSC lines. Scale bars, 200 μm. u, Volcano plot of differentially expressed genes. Red, up-regulated genes in 129XOG2 iPSC; blue, down-regulated genes in 129XOG2 iPSC; R, Pearson correlation coefficient. v, Summary of reprogramming efficiencies. w, iPSC colony number of samples that each FCR small molecule was added in mouse TFR. C, CHIR99021; 6, 616452; F, Forskolin; A, AM580; E, EPZ004777; SR, SR11237; CD, CD3254; V, VPA; CCT, CCT129202; R, R406; D, 5-aza-dC; S, SGC0946; DMH, DMH1; BML, BML-277; AZD, AZD9291 mesylate; CBP, SGC-CBP30; Cit, Citarinostat; TMP, TMP269; WS, WS6; PD, PD0325901. Data are mean ± SD, n = 3 biological replicates. x, iPSC colony number of samples that each FCR small molecule was added in human TFR. Data are mean ± SD, n = 3 biological replicates.

Source data

Extended Data Fig. 3 Profile of FCR process.

a, Read coverage histograms representing the abundance of RNA, ATAC, H3K27ac, H3K4me3, H3K18la, H3K27me3, and H3K9me3 at the genomic loci of Acta2, Gata4, and Oct4. b, Spearman correlations between replicated sequencing samples (heatmaps of correlation matrices), including RNA-seq, ATAC-seq and CUT&Tag (H3K27ac, H3K4me3, H3K18la, H3K9me3 and H3K27me3). Each sequencing sample has 2 biological replicates.

Extended Data Fig. 4 Reprogramming trajectory of FCR.

a, Expression heatmap of 7,201 differentially expressed genes (DEGs, FC > 4 and maximal FPKM > 5), these genes were classified by K-means clustering and fell into 5 stages (left). Typical GO terms and representative genes of each stage (right). P values were calculated by one-tailed hypergeometric test. b, Line charts of representative pluripotency gene expression. c, Expression of Foxa2, Sox17, and Epcam on FCR day 4 as detected by immunofluorescence. Scale bars, 100 μm. d, PCA of normalized gene expression from all FCR samples and CR samples. PC1 and PC2 (left), PC1 and PC3 (right). Reprogramming trajectories were similar but not identical. e, Gene set enrichment analysis (GSEA) of transcriptomes from adjacent stages showed enrichment of the top 10 KEGG pathways (ranked by adjusted P values). P values were calculated by one-tailed hypergeometric test.

Source data

Extended Data Fig. 5 Diapause-like state is a feature unique to FCR.

a, Scatter plots showing gene expression of GO terms and KEGG pathways that significantly changed both in the comparison Diapause ICM/control ICM (x axis) and in D8/D4 (y axis). The gray dots represent all protein-coding genes. The blue dots represent genes in down-regulated terms (DNA replication-related genes: n = 212; Chromosome segregation-related genes: n = 273; Spliceosomal complex-related genes: n = 158; rRNA processing-related genes: n = 176; mRNA processing-related genes: n = 361; Ribosome biogenesis-related genes: n = 247) and the red dots represent genes in up-regulated terms (Lysosome-related genes: n = 120). The boxplots at the margins show the distributions of fold changes. b, Scatter plots showing gene expression of GO terms and KEGG pathways that significantly changed both in the comparison Diapause ICM/control ICM (x axis) and in D8/D4_CF1 (y axis). The gray dots represent all protein-coding genes. The blue dots represent genes in down-regulated terms (Cell cycle-related genes: n = 1,295; DNA replication-related genes: n = 212; Chromosome segregation-related genes: n = 273; RNA splicing-related genes: n = 302; Spliceosomal complex-related genes: n = 158; mRNA processing-related genes: n = 361; rRNA processing-related genes: n = 176; Translation-related genes: n = 489; Ribosome biogenesis-related genes: n = 247) and the red dots represent genes in up-regulated terms (Signaling receptor activity-related genes: n = 230; Lysosome-related genes: n = 120). The boxplots at the margins show the distributions of fold changes. c, Transcription heatmaps of genes involved in DNA replication, chromosome segregation, cell cycle, spliceosomal complex, RNA splicing, mRNA processing, rRNA processing, translation, and ribosome biogenesis. Heatmaps were generated by intra-group normalization of gene expression. In boxplots, boxes represent the median (bold black lines), first and third quartiles, whiskers represent ± 1.5 × interquartile range.

Extended Data Fig. 6 Cell fate transitions during FCR.

a, UMAP visualization of cell clustering of FCR scRNA-seq profiles. b, UMAP visualization of marker gene expression. Blue to grey gradients represent the normalized FPKM of each gene. c, Dot plot showing the expression pattern of fibroblast, mesenchymal cell, epithelial cell, extraembryonic endoderm (XEN), trophectoderm (TE), and pluripotency-related genes in FCR. d, Bar diagrams showing the average regulon activities of representative regulons in different cell types. n = 2 biological replicates. e, Pseudotime order of marker genes along FCR successful trajectory. The black lines represent the mean expression curve of each gene. f, PCA of D12 minus-one RNA-seq data. g-i, Bar diagrams of Oct4, Cdx2 and Sox7 expression in each minus-one RNA-seq datasets, n = 2 biological replicates. j, Expression on heatmap showing the K-means clustering of differentially expressed genes in each minus-one dataset (left), and top KEGG terms of each cluster (right), P value of each KEGG term was calculated by one-tailed hypergeometric test. k, Statistic analysis of FACS results representing EdU incorporation efficiency with the removal of PD0325901. Data are mean ± SD, n = 3 biological replicates. ***P < 0.001 (two-tailed Student’s t test). P value was 0.00058. l, Schematic diagram represents the roles of each small molecule in influencing cell fate determination at the late stage of FCR.

Source data

Extended Data Fig. 7 Dynamic changes of chromatin accessibility, histone modifications, and DNA methylation across FCR.

a, Bar diagrams showing the average ATAC, H3K27ac, H3K4me3, H3K18la, H3K27me3, and H3K9me3 peak abundance along FCR. n = 2 biological replicates. b, Bar diagrams showing the average H3K18la abundance at gained sites in D4 vs D0 (left). GO terms of genes related to these H3K18la_gain sites (right). n = 2 biological replicates. c, Bar diagrams showing the average H3K27me3 abundance at lost sites in D6 vs D0 (left). GO terms of genes related to these H3K27me3_loss sites (right). n = 2 biological replicates. d, Bar diagrams showing the average H3K4me3 abundance at gained sites in iPSC vs D8 (left). GO terms of genes related to these H3K4me3_gain sites (right). n = 2 biological replicates. e, Distribution of low (< 2%), middle (2% - 90%), and high (> 90%) methylation sites on promoters (2-kb around the TSS). Line chart represents average methylation levels. f, Bar diagrams showing the average methylation levels of demethylated promoters in D12 vs D4 (left). GO terms of genes related to these demethylated promoters (right). n = 2 biological replicates. In GO enrichment analysis, P values were calculated by one-tailed hypergeometric test.

Extended Data Fig. 8 Global analysis of chromatin accessibility and histone modifications across FCR.

a, Histograms of ATAC dynamic peak numbers along FCR (left) and TFR (right). Blue: open-to-close loci; red: close-to-open loci. b, Stacked histograms showing the numbers of H3K27ac, H3K4me3, H3K18la, H3K27me3 and H3K9me3 differential peaks and differentially methylated sites (DMSs) along FCR. Blue: down-regulated peaks or DMSs; red: up-regulated peaks or DMSs. c, Venn diagrams of active histone modification peaks along FCR. d, Venn diagrams of repressive histone modification peaks along FCR.

Extended Data Fig. 9 Distributions of histone modifications.

a, Circos (from outer to inner) displays chromosomes (grey), the distributions of protein-coding genes (green), the distributions of H3K9me3 (red), and the distributions of LTR (blue), respectively. Yellow boxes show the representative regions. b, d, f, h, Density plots showing the distance between H3K27ac, H3K18la, H3K4me3, H3K27me3 peaks, and protein-coding genes or LTRs. c, e, g, i, Bar diagrams representing the average distance between H3K27ac, H3K18la, H3K4me3, H3K27me3 peaks and protein-coding genes or LTRs. n = 2 biological replicates. j, Bar diagrams showing the average H3K27ac, H3K18la, H3K4me3 and H3K27me3 abundance in regions of LTR, LINE and SINE.

Extended Data Fig. 10 Schematic diagram.

The development of FCR. In this study, we carried out large-scale chemical screenings, and identified a series of small molecules that can effectively promote chemical reprogramming, and these improvements together amount to the FCR system. Furthermore, we applied genome-wide technologies to profile the FCR process in details, and comprehensively investigated reprogramming trajectory, molecular events, and epigenetic dynamics during FCR, providing rich knowledge about the cell-fate transition.

Supplementary information

Supplementary Information

Supplementary Fig. 1. An example of FACS gating strategy, relative to Fig. 3i.

Reporting Summary

Supplementary Tables

Supplementary Table 1: Key resources. Supplementary Table 2: Small molecule screening data. Supplementary Table 3: Small molecule screening data. Supplementary Table 4: Summary of oligonucleotides. Supplementary Table 5: Summary of shRNAs and sgRNAs.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 6

Statistical 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

Chen, X., Lu, Y., Wang, L. et al. A fast chemical reprogramming system promotes cell identity transition through a diapause-like state. Nat Cell Biol 25, 1146–1156 (2023). https://doi.org/10.1038/s41556-023-01193-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-023-01193-x

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