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.

  • Review Article
  • Published:

Direct cell reprogramming: approaches, mechanisms and progress

Abstract

The reprogramming of somatic cells with defined factors, which converts cells from one lineage into cells of another, has greatly reshaped our traditional views on cell identity and cell fate determination. Direct reprogramming (also known as transdifferentiation) refers to cell fate conversion without transitioning through an intermediary pluripotent state. Given that the number of cell types that can be generated by direct reprogramming is rapidly increasing, it has become a promising strategy to produce functional cells for therapeutic purposes. This Review discusses the evolution of direct reprogramming from a transcription factor-based method to a small-molecule-driven approach, the recent progress in enhancing reprogrammed cell maturation, and the challenges associated with in vivo direct reprogramming for translational applications. It also describes our current understanding of the molecular mechanisms underlying direct reprogramming, including the role of transcription factors, epigenetic modifications, non-coding RNAs, and the function of metabolic reprogramming, and highlights novel insights gained from single-cell omics studies.

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: Principles of indirect and direct reprogramming.
Fig. 2: Direct reprogramming across germ layers.
Fig. 3: Functions of reprogramming factors during direct reprogramming.
Fig. 4: Histone modifications that regulate gene expression during direct reprogramming.
Fig. 5: Metabolic switch during direct reprogramming.
Fig. 6: Single-cell omics in direct reprogramming.

Similar content being viewed by others

References

  1. Waddington, C. H. The Strategy of the Genes. A Discussion of Some Aspects of Theoretical Biology. With an Appendix by H. Kacser (George Allen & Unwin, Ltd., 1957).

  2. Davis, R. L., Weintraub, H. & Lassar, A. B. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell 51, 987–1000 (1987). Davis et al. demonstrated, for the first time, that the overexpression of one transcription factor could rewrite cell fate in vitro.

    Article  CAS  PubMed  Google Scholar 

  3. 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 

  4. Yamanaka, S. Induced pluripotent stem cells: past, present, and future. Cell Stem Cell 10, 678–684 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Buganim, Y., Faddah, D. A. & Jaenisch, R. Mechanisms and models of somatic cell reprogramming. Nat. Rev. Genet. 14, 427–439 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  7. Smith, Z. D., Sindhu, C. & Meissner, A. Molecular features of cellular reprogramming and development. Nat. Rev. Mol. Cell Biol. 17, 139–154 (2016).

    CAS  PubMed  Google Scholar 

  8. Srivastava, D. & DeWitt, N. In vivo cellular reprogramming: the next generation. Cell 166, 1386–1396 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Jorstad, N. L. et al. Stimulation of functional neuronal regeneration from Müller glia in adult mice. Nature 548, 103–107 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Li, H. & Chen, G. In vivo reprogramming for CNS repair: regenerating neurons endogenous glial cells. Neuron 91, 728–738 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Qian, L. et al. In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature 485, 593–598 (2012). Qian et al. demonstrated the feasibility of using in vivo direct reprogramming for heart repair.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Stone, N. R. et al. Context-specific transcription factor functions regulate epigenomic and transcriptional dynamics during cardiac reprogramming. Cell Stem Cell 25, 87–102.e9 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kulessa, H., Frampton, J. & Graf, T. GATA-1 reprograms avian myelomonocytic cell lines into eosinophils, thromboblasts, and erythroblasts. Genes Dev. 9, 1250–1262 (1995).

    Article  CAS  PubMed  Google Scholar 

  15. Xie, H., Ye, M., Feng, R. & Graf, T. Stepwise reprogramming of B cells into macrophages. Cell 117, 663–676 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Laiosa, C. V., Stadtfeld, M., Xie, H., de Andres-Aguayo, L. & Graf, T. Reprogramming of committed T cell progenitors to macrophages and dendritic cells by C/EBPα and PU.1 transcription factors. Immunity 25, 731–744 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Zhou, Q., Brown, J., Kanarek, A., Rajagopal, J. & Melton, D. A. In vivo reprogramming of adult pancreatic exocrine cells to β-cells. Nature 455, 627–632 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ieda, M. et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell 142, 375–386 (2010). Idea et al. identified reprogramming factors that could reprogramme mouse cardiac fibroblasts to cardiomyocyte-like cells in vitro.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Song, K. et al. Heart repair by reprogramming non-myocytes with cardiac transcription factors. Nature 485, 599–604 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Vierbuchen, T. et al. Direct conversion of fibroblasts to functional neurons by defined factors. Nature 463, 1035–1041 (2010). Vierbuchen et al. identified a combination of three factors to directly convert mouse fibroblasts into functional neurons in vitro.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Sekiya, S. & Suzuki, A. Direct conversion of mouse fibroblasts to hepatocyte-like cells by defined factors. Nature 475, 390–393 (2011).

    Article  CAS  PubMed  Google Scholar 

  22. Huang, P. et al. Induction of functional hepatocyte-like cells from mouse fibroblasts by defined factors. Nature 475, 386–389 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Jayawardena, T. M. et al. MicroRNA-mediated in vitro and in vivo direct reprogramming of cardiac fibroblasts to cardiomyocytes. Circ. Res. 110, 1465–1473 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Jayawardena, T. M. et al. MicroRNA induced cardiac reprogramming in vivo. Circ. Res. 116, 418–424 (2015).

    Article  CAS  PubMed  Google Scholar 

  26. Xu, J., Du, Y. & Deng, H. Direct lineage reprogramming: strategies, mechanisms, and applications. Cell Stem Cell 16, 119–134 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Takahashi, J. Strategies for bringing stem cell-derived dopamine neurons to the clinic: the Kyoto trial. in Progress in Brain Research 230, 213–226 (Elsevier B.V., 2017).

  28. Barker, R. A., Parmar, M., Studer, L. & Takahashi, J. Human trials of stem cell-derived dopamine neurons for Parkinson’s disease: dawn of a new era. Cell Stem Cell 21, 569–573 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Zarbin, M., Sugino, I. & Townes-Anderson, E. Concise review: update on retinal pigment epithelium transplantation for age-related macular degeneration. Stem Cell Transl. Med. 8, 466–477 (2019).

    Article  Google Scholar 

  30. Blau, H. M. & Daley, G. Q. Stem cells in the treatment of disease. N. Engl. J. Med. 380, 1748–1760 (2019).

    Article  CAS  PubMed  Google Scholar 

  31. Li, H. & Chen, G. In vivo reprogramming for CNS repair: regenerating neurons from endogenous glial cells. Neuron 91, 728–738 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Jayawardena, T. M. et al. MicroRNA induced cardiac reprogramming in vivo evidence for mature cardiac myocytes and improved cardiac function. Circ. Res. 116, 418–424 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Niu, W. et al. In vivo reprogramming of astrocytes to neuroblasts in the adult brain. Nat. Cell Biol. 15, 1164–1175 (2013).

    Article  CAS  PubMed  Google Scholar 

  34. Grande, A. et al. Environmental impact on direct neuronal reprogramming in vivo in the adult brain. Nat. Commun. 4, 2373 (2013).

    Article  PubMed  Google Scholar 

  35. Guo, Z. et al. In vivo direct reprogramming of reactive glial cells into functional neurons after brain injury and in an Alzheimer’s disease model. Cell Stem Cell 14, 188–202 (2014).

    Article  CAS  PubMed  Google Scholar 

  36. Torper, O. et al. Generation of induced neurons via direct conversion in vivo. Proc. Natl Acad. Sci. USA 110, 7038–7043 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Heinrich, C. et al. Sox2-mediated conversion of NG2 glia into induced neurons in the injured adult cerebral cortex. Stem Cell Rep. 3, 1000–1014 (2014).

    Article  CAS  Google Scholar 

  38. Su, Z., Niu, W., Liu, M. L., Zou, Y. & Zhang, C. L. In vivo conversion of astrocytes to neurons in the injured adult spinal cord. Nat. Commun. 5, 3338 (2014).

    Article  PubMed  CAS  Google Scholar 

  39. Song, G. et al. Direct reprogramming of hepatic myofibroblasts into hepatocytes in vivo attenuates liver fibrosis. Cell Stem Cell 18, 797–808 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Yao, K. et al. Restoration of vision after de novo genesis of rod photoreceptors in mammalian retinas. Nature 560, 484–488 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Fu, J. D. & Srivastava, D. Direct reprogramming of fibroblasts into cardiomyocytes for cardiac regenerative medicine. Circ. J. 79, 245–254 (2015).

    Article  PubMed  Google Scholar 

  42. Gascón, S., Masserdotti, G., Russo, G. L. & Götz, M. Direct neuronal reprogramming: achievements, hurdles, and new roads to success. Cell Stem Cell 21, 18–34 (2017).

    Article  PubMed  CAS  Google Scholar 

  43. Li, W. et al. Long-term persistence and development of induced pancreatic beta cells generated by lineage conversion of acinar cells. Nat. Biotechnol. 32, 1223–1230 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Thorel, F. et al. Conversion of adult pancreatic α-cells to B-cells after extreme B-cell loss. Nature 464, 1149–1154 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Humeres, C. & Frangogiannis, N. G. Fibroblasts in the infarcted, remodeling, and failing heart. JACC Basic. Transl. Sci. 4, 449–467 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Jin, Y. et al. Three-dimensional brain-like microenvironments facilitate the direct reprogramming of fibroblasts into therapeutic neurons. Nat. Biomed. Eng. 2, 522–539 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Li, Y. et al. Tissue-engineered 3-dimensional (3D) microenvironment enhances the direct reprogramming of fibroblasts into cardiomyocytes by microRNAs. Sci. Rep. 6, 38815 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Magnusson, J. P. et al. A latent neurogenic program in astrocytes regulated by Notch signaling in the mouse. Science 346, 237–241 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Hu, X. et al. Region-restrict astrocytes exhibit heterogeneous susceptibility to neuronal reprogramming. Stem Cell Rep. 12, 290–304 (2019).

    Article  CAS  Google Scholar 

  50. Buffo, A. et al. Origin and progeny of reactive gliosis: a source of multipotent cells in the injured brain. Proc. Natl Acad. Sci. USA 105, 3581–3586 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kang, S. H., Fukaya, M., Yang, J. K., Rothstein, J. D. & Bergles, D. E. NG2+ CNS glial progenitors remain committed to the oligodendrocyte lineage in postnatal life and following neurodegeneration. Neuron 68, 668–681 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. De La Rossa, A. et al. In vivo reprogramming of circuit connectivity in postmitotic neocortical neurons. Nat. Neurosci. 16, 193–200 (2013).

    Article  PubMed  CAS  Google Scholar 

  53. Rouaux, C. & Arlotta, P. Direct lineage reprogramming of post-mitotic callosal neurons into corticofugal neurons in vivo. Nat. Cell Biol. 15, 214–221 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Xiao, X. et al. Endogenous reprogramming of alpha cells into beta cells, induced by viral gene therapy, reverses autoimmune diabetes. Cell Stem Cell 22, 78–90.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Collombat, P. et al. The ectopic expression of Pax4 in the mouse pancreas converts progenitor cells into α and subsequently β cells. Cell 138, 449–462 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Bramswig, N. C. et al. Epigenomic plasticity enables human pancreatic α to β cell reprogramming. J. Clin. Invest. 123, 1275–1284 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Liu, Y. et al. Ascl1 converts dorsal midbrain astrocytes into functional neurons in vivo. J. Neurosci. 35, 9336–9355 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Colasante, G. et al. Rapid conversion of fibroblasts into functional forebrain GABAergic interneurons by direct genetic reprogramming. Cell Stem Cell 17, 719–734 (2015).

    Article  CAS  PubMed  Google Scholar 

  59. Li, W. et al. In vivo reprogramming of pancreatic acinar cells to three islet endocrine subtypes. eLife 3, 1846 (2014).

    Google Scholar 

  60. Nam, Y. J. et al. Induction of diverse cardiac cell types by reprogramming fibroblasts with cardiac transcription factors. Development 141, 4267–4278 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Miyamoto, K. et al. Direct in vivo reprogramming with sendai virus vectors improves cardiac function after myocardial infarction. Cell Stem Cell 22, 91–103.e5 (2018).

    Article  CAS  PubMed  Google Scholar 

  62. Lee, K. et al. Peptide-enhanced mRNA transfection in cultured mouse cardiac fibroblasts and direct reprogramming towards cardiomyocyte-like cells. Int. J. Nanomed. 10, 1841–1854 (2015).

    CAS  Google Scholar 

  63. Chakraborty, S. et al. A CRISPR/Cas9-based system for reprogramming cell lineage specification. Stem Cell Rep. 3, 940–947 (2014).

    Article  CAS  Google Scholar 

  64. Liu, Y. et al. CRISPR activation screens systematically identify factors that drive neuronal fate and reprogramming. Cell Stem Cell 23, 758–771.e8 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Chang, Y. et al. Efficient in vivo direct conversion of fibroblasts into cardiomyocytes using a nanoparticle-based gene carrier. Biomaterials 192, 500–509 (2019).

    Article  CAS  PubMed  Google Scholar 

  66. Fu, Y. et al. Direct reprogramming of mouse fibroblasts into cardiomyocytes with chemical cocktails. Cell Res. 25, 1013–1024 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Park, G. et al. Conversion of mouse fibroblasts into cardiomyocyte-like cells using small molecule treatments. Biomaterials 54, 201–212 (2015).

    Article  CAS  PubMed  Google Scholar 

  68. Cao, N. et al. Conversion of human fibroblasts into functional cardiomyocytes by small molecules. Science 352, 1216–1220 (2016).

    Article  CAS  PubMed  Google Scholar 

  69. Wang, Y. et al. Chemical conversion of mouse fibroblasts into functional dopaminergic neurons. Exp. Cell Res. 347, 283–292 (2016).

    Article  CAS  PubMed  Google Scholar 

  70. Dai, P., Harada, Y. & Takamatsu, T. Highly efficient direct conversion of human fibroblasts to neuronal cells by chemical compounds. J. Clin. Biochem. Nutr. 56, 166–170 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Li, X. et al. Small-molecule-driven direct reprogramming of mouse fibroblasts into functional neurons. Cell Stem Cell 17, 195–203 (2015).

    Article  CAS  PubMed  Google Scholar 

  72. Zhang, L. et al. Small molecules efficiently reprogram human astroglial cells into functional neurons. Cell Stem Cell 17, 735–747 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Yu, C., Liu, K., Tang, S. & Ding, S. Chemical approaches to cell reprogramming. Curr. Opin. Genet. Dev. 28, 50–56 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Li, J. & Mooney, D. J. Designing hydrogels for controlled drug delivery. Nat. Rev. Mater. 1, 16071 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Liu, Z. et al. Single-cell transcriptomics reconstructs fate conversion from fibroblast to cardiomyocyte. Nature 551, 100–104 (2017). Liu et al. constructed the routes of iCM formation using single-cell RNA-seq and discovered the critical role of splicing factor PTBP1 during murine cardiac reprogramming.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Zhou, Y. et al. Single-cell transcriptomic analyses of cell fate transitions during human cardiac reprogramming. Cell Stem Cell 25, 149–164.e9 (2019). Zhou et al. revealed an early decision point at which cells either continue reprogramming or regress toward the original fibroblast state during human cardiac reprogramming.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Treutlein, B. et al. Dissecting direct reprogramming from fibroblast to neuron using single-cell RNA-seq. Nature 534, 391–395 (2016). Treutlein et al. described the existence of an alternative reprogramming route in murine neuronal reprogramming using single-cell RNA-seq.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Richards, D. J. et al. Human cardiac organoids for the modelling of myocardial infarction and drug cardiotoxicity. Nat. Biomed. Eng. 4, 446–462 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Noor, N. et al. 3D printing of personalized thick and perfusable cardiac patches and hearts. Adv. Sci. 6, 1900344 (2019).

    Article  CAS  Google Scholar 

  80. Zhou, Y. et al. Bmi1 is a key epigenetic barrier to direct cardiac reprogramming. Cell Stem Cell 18, 382–395 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Qian, H. et al. Reversing a model of Parkinson’s disease with in situ converted nigral neurons. Nature 582, 550–556 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Trokovic, R., Weltner, J., Noisa, P., Raivio, T. & Otonkoski, T. Combined negative effect of donor age and time in culture on the reprogramming efficiency into induced pluripotent stem cells. Stem Cell Res. 15, 254–262 (2015).

    Article  PubMed  Google Scholar 

  83. Lo Sardo, V. et al. Influence of donor age on induced pluripotent stem cells. Nat. Biotechnol. 35, 69–74 (2017).

    Article  CAS  PubMed  Google Scholar 

  84. 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 

  85. Iwafuchi-Doi, M. & Zaret, K. S. Pioneer transcription factors in cell reprogramming. Genes Dev. 28, 2679–2692 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Cirillo, L. A. et al. Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA-4. Mol. Cell 9, 279–289 (2002).

    Article  CAS  PubMed  Google Scholar 

  87. Wapinski, O. L. et al. Hierarchical mechanisms for direct reprogramming of fibroblasts to neurons. Cell 155, 621–635 (2013). Wapinski et al. describe the role of Ascl1 as an ‘on-target’ Pioneer factor and its association to the ‘trivalent’ chromatin state in murine neuronal reprogramming.

    Article  CAS  PubMed  Google Scholar 

  88. Soufi, A., Donahue, G. & Zaret, K. S. Facilitators and impediments of the pluripotency reprogramming factors’ initial engagement with the genome. Cell 151, 994–1004 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Chanda, S. et al. Generation of induced neuronal cells by the single reprogramming factor ASCL1. Stem Cell Rep. 3, 282–296 (2014).

    Article  CAS  Google Scholar 

  90. Hashimoto, H. et al. Cardiac reprogramming factors synergistically activate genome-wide cardiogenic stage-specific enhancers. Cell Stem Cell 25, 69–86.e5 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Zhou, H., Dickson, M. E., Kim, M. S., Bassel-Duby, R. & Olson, E. N. Akt1/protein kinase B enhances transcriptional reprogramming of fibroblasts to functional cardiomyocytes. Proc. Natl Acad. Sci. USA 112, 11864–11869 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Wang, L. et al. Improved generation of induced cardiomyocytes using a polycistronic construct expressing optimal ratio of Gata4, Mef2c and Tbx5. J. Vis. Exp. 105, 53426 (2015).

    Google Scholar 

  93. Wang, L. et al. Stoichiometry of Gata4, Mef2c, and Tbx5 influences the efficiency and quality of induced cardiac myocyte reprogramming. Circ. Res. 116, 237–244 (2015).

    Article  PubMed  CAS  Google Scholar 

  94. Mathison, M. et al. ‘Triplet’ polycistronic vectors encoding Gata4, Mef2c, and Tbx5 enhances postinfarct ventricular functional improvement compared with singlet vectors. J. Thorac. Cardiovasc. Surg. 148, 1656–1664.e2 (2014).

    Article  CAS  PubMed  Google Scholar 

  95. Horisawa, K. et al. The dynamics of transcriptional activation by hepatic reprogramming factors. Mol. Cell 79, 660–676.e8 (2020).

    Article  CAS  PubMed  Google Scholar 

  96. Wapinski, O. L. et al. Rapid chromatin switch in the direct reprogramming of fibroblasts to neurons. Cell Rep. 20, 3236–3247 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Bannister, A. J. & Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 21, 381–395 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Liu, Z. et al. Re-patterning of H3K27me3, H3K4me3 and DNA methylation during fibroblast conversion into induced cardiomyocytes. Stem Cell Res. 16, 507–518 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Barbagiovanni, G. et al. KMT2B is selectively required for neuronal transdifferentiation, and its loss exposes dystonia candidate genes. Cell Rep. 25, 988–1001 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Benayoun, B. A. et al. H3K4me3 breadth is linked to cell identity and transcriptional consistency. Cell 158, 673–688 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Ezhkova, E. et al. EZH1 and EZH2 cogovern histone H3K27 trimethylation and are essential for hair follicle homeostasis and wound repair. Genes Dev. 25, 485–498 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Dal-Pra, S., Hodgkinson, C. P., Mirotsou, M., Kirste, I. & Dzau, V. J. Demethylation of H3K27 is essential for the induction of direct cardiac reprogramming by MIR Combo. Circulation Res. 120, 1403–1413 (2017).

    Article  CAS  PubMed  Google Scholar 

  104. Becker, J. S., Nicetto, D. & Zaret, K. S. H3K9me3-dependent heterochromatin: barrier to cell fate changes. Trends Genet. 32, 29–41 (2016).

    Article  CAS  PubMed  Google Scholar 

  105. Huang, P. et al. Direct reprogramming of human fibroblasts to functional and expandable hepatocytes. Cell Stem Cell 14, 370–384 (2014).

    Article  CAS  PubMed  Google Scholar 

  106. Becker, J. S. et al. Genomic and proteomic resolution of heterochromatin and its restriction of alternate fate genes. Mol. Cell 68, 1023–1037.e15 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Hirai, H. & Kikyo, N. Inhibitors of suppressive histone modification promote direct reprogramming of fibroblasts to cardiomyocyte-like cells. Cardiovasc. Res. 102, 188–190 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Rhee, C. et al. Mechanisms of transcription factor-mediated direct reprogramming of mouse embryonic stem cells to trophoblast stem-like cells. Nucleic Acids Res. 45, 10103–10114 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Cao, J. & Yan, Q. Histone ubiquitination and deubiquitination in transcription, DNA damage response, and cancer. Front. Oncol. 2, 26 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  111. Vastenhouw, N. L. & Schier, A. F. Bivalent histone modifications in early embryogenesis. Curr. Opin. Cell Biol. 24, 374–386 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Bernstein, B. E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    Article  CAS  PubMed  Google Scholar 

  113. Santoro, S. W. & Dulac, C. Histone variants and cellular plasticity. Trends Genet. 31, 516–527 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Henikoff, S. & Smith, M. M. Histone variants and epigenetics. Cold Spring Harb. Perspect. Biol. 7, a019364 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Gaspar-Maia, A. et al. MacroH2A histone variants act as a barrier upon reprogramming towards pluripotency. Nat. Commun. 4, 1565 (2013).

    Article  PubMed  CAS  Google Scholar 

  116. Dong, F. et al. Dynamic changes in occupancy of histone variant H2a.Z during induced somatic cell reprogramming. Stem Cells Int. 2016, 3162363 (2016).

    Article  PubMed  CAS  Google Scholar 

  117. Fang, H. T. et al. Global H3.3 dynamic deposition defines its bimodal role in cell fate transition. Nat. Commun. 9, 1537 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  118. Cheng, H. et al. Reprogramming mouse fibroblasts into engraftable myeloerythroid and lymphoid progenitors. Nat. Commun. 7, 13396 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Luo, C. et al. Global DNA methylation remodeling during direct reprogramming of fibroblasts to neurons. eLife 8, e40197 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Luginbühl, J., Sivaraman, D. M. & Shin, J. W. The essentiality of non-coding RNAs in cell reprogramming. Noncoding RNA Res. 2, 74–82 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  121. Muraoka, N. et al. MiR-133 promotes cardiac reprogramming by directly repressing Snai1 and silencing fibroblast signatures. EMBO J. 33, 1565–1581 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Abernathy, D. G. et al. MicroRNAs induce a permissive chromatin environment that enables neuronal subtype-specific reprogramming of adult human fibroblasts. Cell Stem Cell 21, 332–348.e9 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Lee, S. W., Oh, Y. M., Lu, Y. L., Kim, W. K. & Yoo, A. S. MicroRNAs overcome cell fate barrier by reducing EZH2-controlled REST stability during neuronal conversion of human adult fibroblasts. Dev. Cell 46, 73–84.e7 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Ballas, N., Grunseich, C., Lu, D. D., Speh, J. C. & Mandel, G. REST and its corepressors mediate plasticity of neuronal gene chromatin throughout neurogenesis. Cell 121, 645–657 (2005).

    Article  CAS  PubMed  Google Scholar 

  126. Conaco, C., Otto, S., Han, J. J. & Mandel, G. Reciprocal actions of REST and a microRNA promote neuronal identity. Proc. Natl Acad. Sci. USA 103, 2422–2427 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Cusanovich, D. A. et al. A single-cell atlas of in vivo mammalian chromatin accessibility. Cell 174, 1309–1324.e18 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Adachi, K. et al. Esrrb unlocks silenced enhancers for reprogramming to naive pluripotency. Cell Stem Cell 23, 266–275.e6 (2018).

    Article  CAS  PubMed  Google Scholar 

  129. Ang, C. E. et al. The novel lncRNA lnc-NR2F1 is pro-neurogenic and mutated in human neurodevelopmental disorders. eLife 8, e41770 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  130. Loewer, S. et al. Large intergenic non-coding RNA-RoR modulates reprogramming of human induced pluripotent stem cells. Nat. Genet. 42, 1113–1117 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Wang, Y. et al. Endogenous miRNA sponge lincRNA-RoR regulates Oct4, Nanog, and Sox2 in human embryonic stem cell self-renewal. Dev. Cell 25, 69–80 (2013).

    Article  CAS  PubMed  Google Scholar 

  132. Bao, X. et al. The p53-induced lincRNA-p21 derails somatic cell reprogramming by sustaining H3K9me3 and CpG methylation at pluripotency gene promoters. Cell Res. 25, 80–92 (2015).

    Article  CAS  PubMed  Google Scholar 

  133. Guttman, M. et al. lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature 477, 295–300 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Ng, S.-Y., Johnson, R. & Stanton, L. W. Human long non-coding RNAs promote pluripotency and neuronal differentiation by association with chromatin modifiers and transcription factors. EMBO J. 31, 522–533 (2012).

    Article  CAS  PubMed  Google Scholar 

  135. Jain, A. K. et al. LncPRESS1 Is a p53-regulated LncRNA that safeguards pluripotency by disrupting SIRT6-mediated de-acetylation of histone H3K56. Mol. Cell 64, 967–981 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Dinger, M. E. et al. Long noncoding RNAs in mouse embryonic stem cell pluripotency and differentiation. Genome Res. 18, 1433–1445 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Zheng, X. et al. Metabolic reprogramming during neuronal differentiation from aerobic glycolysis to neuronal oxidative phosphorylation. eLife 5, e13374 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  138. Cliff, T. S. & Dalton, S. Metabolic switching and cell fate decisions: implications for pluripotency, reprogramming and development. Curr. Opin. Genet. Dev. 46, 44–49 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Mathieu, J. & Ruohola-Baker, H. Metabolic remodeling during the loss and acquisition of pluripotency. Development 144, 541–551 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Gascón, S. et al. Identification and successful negotiation of a metabolic checkpoint in direct neuronal reprogramming. Cell Stem Cell 18, 396–409 (2016). Gascón et al. revealed a critical metabolic checkpoint important for successful murine neuronal reprogramming in vivo and in vitro.

    Article  PubMed  CAS  Google Scholar 

  141. Magistretti, P. J. & Allaman, I. A cellular perspective on brain energy metabolism and functional imaging. Neuron 86, 883–901 (2015).

    Article  CAS  PubMed  Google Scholar 

  142. Lopaschuk, G. D. & Jaswal, J. S. Energy metabolic phenotype of the cardiomyocyte during development, differentiation, and postnatal maturation. J. Cardiovasc. Pharmacol. 56, 130–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  143. Zhou, Y. et al. Comparative gene expression analyses reveal distinct molecular signatures between differentially reprogrammed cardiomyocytes. Cell Rep. 20, 3014–3024 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Li, X. et al. Mitochondrial ROS, uncoupled from ATP synthesis, determine endothelial activation for both physiological recruitment of patrolling cells and pathological recruitment of inflammatory cells. Can. J. Physiol. Pharmacol. 95, 247–252 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  145. Finkel, T. Signal transduction by mitochondrial oxidants. J. Biol. Chem. 287, 4434–4440 (2012).

    Article  CAS  PubMed  Google Scholar 

  146. West, A. P. et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature 472, 476–480 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Wang, X. et al. Selenium augments microRNA directed reprogramming of fibroblasts to cardiomyocytes via nanog. Sci. Rep. 6, 23017 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Tang, F. et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat. Methods 6, 377–382 (2009).

    Article  CAS  PubMed  Google Scholar 

  149. Hwang, B., Lee, J. H. & Bang, D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp. Mol. Med. 50, 96 (2018).

    Article  CAS  PubMed Central  Google Scholar 

  150. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Zheng, G. X. Y. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Kiselev, V. Y., Andrews, T. S. & Hemberg, M. Challenges in unsupervised clustering of single-cell RNA-seq data. Nat. Rev. Genet. 20, 273–282 (2019).

    Article  CAS  PubMed  Google Scholar 

  153. Mohamed, T. M. A. et al. Regulation of cell cycle to stimulate adult cardiomyocyte proliferation and cardiac regeneration. Cell 173, 104–116.e12 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Xin, M. et al. Hippo pathway effector Yap promotes cardiac regeneration. Proc. Natl Acad. Sci. USA 110, 13839–13844 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Leach, J. P. et al. Hippo pathway deficiency reverses systolic heart failure after infarction. Nature 550, 260–264 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  156. Heallen, T. et al. Hippo pathway inhibits wnt signaling to restrain cardiomyocyte proliferation and heart size. Science 332, 458–461 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Trapnell, C. Defining cell types and states with single-cell genomics. Genome Res. 25, 1491–1498 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Saelens, W., Cannoodt, R., Todorov, H. & Saeys, Y. A comparison of single-cell trajectory inference methods. Nat. Biotechnol. 37, 547–554 (2019). Saelens et al. comprehensively evaluated the performance of 45 single-cell trajectory inference methods on 110 real and 229 synthetic datasets and provide guidelines for method selection in direct reprogramming.

    Article  CAS  PubMed  Google Scholar 

  159. 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  CAS  PubMed  PubMed Central  Google Scholar 

  160. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  161. Rozenblatt-Rosen, O., Stubbington, M. J. T., Regev, A. & Teichmann, S. A. The human cell atlas: from vision to reality. Nature 550, 451–453 (2017).

    Article  CAS  PubMed  Google Scholar 

  162. Stuart, T. & Satija, R. Integrative single-cell analysis. Nat. Rev. Genet. 20, 257–272 (2019).

    Article  CAS  PubMed  Google Scholar 

  163. Stegle, O., Teichmann, S. A. & Marioni, J. C. Computational and analytical challenges in single-cell transcriptomics. Nat. Rev. Genet. 16, 133–145 (2015).

    Article  CAS  PubMed  Google Scholar 

  164. Grün, D. et al. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature 525, 251–255 (2015).

    Article  PubMed  CAS  Google Scholar 

  165. Jia, C. et al. Accounting for technical noise in differential expression analysis of single-cell RNA sequencing data. Nucleic Acids Res. 45, 10978–10988 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Mahata, B. et al. Single-cell RNA sequencing reveals T helper cells synthesizing steroids De Novo to contribute to immune homeostasis. Cell Rep. 7, 1130–1142 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Treutlein, B. et al. Reconstructing lineage hierarchies of the distal lung epithelium using single-cell RNA-seq. Nature 509, 371–375 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Buenrostro, J. D. et al. Integrated single-cell analysis maps the continuous regulatory landscape of human hematopoietic differentiation. Cell 173, 1535–1548.e16 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Nagano, T. et al. Cell-cycle dynamics of chromosomal organization at single-cell resolution. Nature 547, 61–67 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Wang, Q. et al. CoBATCH for high-throughput single-cell epigenomic profiling. Mol. Cell 76, 206–216.e7 (2019).

    Article  CAS  PubMed  Google Scholar 

  171. Cao, J. et al. Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science 361, 1380–1385 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Welch, J. D. et al. Single-cell multi-omic integration compares and contrasts features of brain cell identity. Cell 177, 1873–1887.e17 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Welch, J. D., Hartemink, A. J. & Prins, J. F. MATCHER: manifold alignment reveals correspondence between single cell transcriptome and epigenome dynamics. Genome Biol. 18, 138 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  174. Cheow, L. F. et al. Single-cell multimodal profiling reveals cellular epigenetic heterogeneity. Nat. Methods 13, 833–836 (2016).

    Article  CAS  PubMed  Google Scholar 

  175. Eraslan, G., Avsec, Ž., Gagneur, J. & Theis, F. J. Deep learning: new computational modelling techniques for genomics. Nat. Rev. Genet. 20, 389–403 (2019).

    Article  CAS  PubMed  Google Scholar 

  176. Zhou, H. et al. ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression. Genes Dev. 31, 1770–1783 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Tang, Y., Liu, M. L., Zang, T. & Zhang, C. L. Direct reprogramming rather than iPSC-based reprogramming maintains aging hallmarks in human motor neurons. Front. Mol. Neurosci. 10, 359 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  178. Fulton, D. L. et al. TFCat: the curated catalog of mouse and human transcription factors. Genome Biol. 10, R29 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  179. Rackham, O. J. L. et al. A predictive computational framework for direct reprogramming between human cell types. Nat. Genet. 48, 331–335 (2016).

    Article  CAS  PubMed  Google Scholar 

  180. D’Alessio, A. C. et al. A systematic approach to identify candidate transcription factors that control cell identity. Stem Cell Rep. 5, 763–775 (2015).

    Article  CAS  Google Scholar 

  181. Forrest, A. R. R. et al. A promoter-level mammalian expression atlas. Nature 507, 462–470 (2014).

    Article  CAS  PubMed  Google Scholar 

  182. Franceschini, A. et al. STRING v9.1: protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Res. 41, D808–D815 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  183. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517, 583–588 (2014).

    PubMed  PubMed Central  Google Scholar 

  184. Karemaker, I. D. & Vermeulen, M. Single-cell DNA methylation profiling: technologies and biological applications. Trends Biotechnol. 36, 952–965 (2018).

    Article  CAS  PubMed  Google Scholar 

  185. 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 

  186. Pott, S. & Lieb, J. D. Single-cell ATAC-seq: strength in numbers. Genome Biol. 16, 172 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  187. Li, G. et al. Joint profiling of DNA methylation and chromatin architecture in single cells. Nat. Methods 16, 991–993 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Ramani, V. et al. Massively multiplex single-cell Hi-C. Nat. Methods 14, 263–266 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Zhu, C. et al. An ultra high-throughput method for single-cell joint analysis of open chromatin and transcriptome. Nat. Struct. Mol. Biol. 26, 1063–1070 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Lee, D. S. et al. Simultaneous profiling of 3D genome structure and DNA methylation in single human cells. Nat. Methods 16, 999–1006 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Spektor, R., Tippens, N. D., Mimoso, C. A. & Soloway, P. D. Methyl-ATAC-seq measures DNA methylation at accessible chromatin. Genome Res. 29, 969–977 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We sincerely apologize to those whose work may not have been cited owing to space constraints. J.L. is supported by NIH/NHLBI R01HL139880, R01HL139976; L.Q. is supported by AHA 18TPA34180058, NIH/NHLBI R01HL128331, R01HL144551.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Li Qian.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Molecular Cell Biology thanks the anonymous reviewers 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.

Related links

FANTOM5: http://fantom.gsc.riken.jp/5/

Mogrify: http://www.mogrify.net/

STRING: https://string-db.org/

Supplementary information

Glossary

Myoblasts

The embryonic precursors of myocytes (muscle cells).

Eosinophils

A type of white blood cell containing granules that could be intensively stained with eosin.

Embryonic germ layer

A layer of cells that form at the early stages of embryonic development. The three embryonic germ layers are the endoderm, ectoderm and mesoderm. Cells in each germ layer interact with each other and differentiate to form tissues and embryonic organs.

Autologous cell therapies

A novel therapeutic intervention that utilizes patients’ own cells to obtain therapeutic cells through ex vivo differentiation or reprogramming.

Microenvironment

The surrounding environment of a cell that contains chemical and physical signals that directly or indirectly regulate cellular behaviour.

Macroglial cells

The non-neuronal cells that provide support and protection for neurons.

Sendai virus

A single strand, negative-sense RNA virus that has a large capacity for gene expression and a wide host range.

Single guide RNA

RNA molecule that contains a short sequence complementary to the target DNA sequence and is used to direct Cas9 endonuclease to target loci.

Hydrogel

A network of polymer chains that are hydrophilic and has been extensively studied as a scaffold for in vivo drug delivery.

Poised enhancers

A subclass of enhancers enriched for both the active and the repressive histone marks. In pluripotent cells, these poised enhancers are located near key early developmental genes and are primed to activate target gene expression upon the right environmental cues.

DNA methyltransferase Dnmt3a

Dnmt3a is an enzyme that catalyses the addition of methyl groups to unmethylated DNA at specific CpG sites.

Epithelial−mesenchymal transition

The process where polarized epithelial cells are transformed into mobile and extracellular matrix-secreting mesenchymal cells.

Oxidative phosphorylation

(OxPhos). A process in which ATP is produced because of electron transfer in mitochondria. OxPhos is the main energy source for cells like neurons, cardiomyocytes or muscle-skeletal cells.

Reactive oxygen species

(ROS). A natural by-product of the electron transport chain during the oxidation of glucose. ROS can act as signalling molecules; high levels of ROS can lead to oxidative stress in a cell.

Glycolysis

A metabolic pathway that converts glucose to pyruvate and produces two ATPs. In proliferating cells, glycolysis is a major resource for energy and macromolecules for biosynthesis.

Microfluidics

The precise control and manipulation of fluids at a small scale.

RNA velocity

The time derivative of the gene expression state. It can be used to predict the future state of individual cells on a timescale of hours.

CpG islands

Short interspersed DNA sequences that are 1,000 base pairs on average and show an unusually elevated level of CpG dinucleotides. Most of the CpG islands are found at gene promoters.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, H., Yang, Y., Liu, J. et al. Direct cell reprogramming: approaches, mechanisms and progress. Nat Rev Mol Cell Biol 22, 410–424 (2021). https://doi.org/10.1038/s41580-021-00335-z

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-021-00335-z

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