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.

A comprehensive library of human transcription factors for cell fate engineering

Abstract

Human pluripotent stem cells (hPSCs) offer an unprecedented opportunity to model diverse cell types and tissues. To enable systematic exploration of the programming landscape mediated by transcription factors (TFs), we present the Human TFome, a comprehensive library containing 1,564 TF genes and 1,732 TF splice isoforms. By screening the library in three hPSC lines, we discovered 290 TFs, including 241 that were previously unreported, that induce differentiation in 4 days without alteration of external soluble or biomechanical cues. We used four of the hits to program hPSCs into neurons, fibroblasts, oligodendrocytes and vascular endothelial-like cells that have molecular and functional similarity to primary cells. Our cell-autonomous approach enabled parallel programming of hPSCs into multiple cell types simultaneously. We also demonstrated orthogonal programming by including oligodendrocyte-inducible hPSCs with unmodified hPSCs to generate cerebral organoids, which expedited in situ myelination. Large-scale combinatorial screening of the Human TFome will complement other strategies for cell engineering based on developmental biology and computational systems biology.

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: Creation of the Human TFome expression library and its application for cell fate engineering.
Fig. 2: ATOH1 induces neurons and NKX3-1 induces fibroblasts in lineage-independent media.
Fig. 3: ETV2 isoform 2 induces vascular endothelial-like cells that form perfusable blood vessels in vivo.
Fig. 4: Parallel programming enables simultaneous differentiation of multiple cell types in the same dish.
Fig. 5: SOX9 induces oligodendrocytes that engraft and form compact myelin in vivo and in cerebral organoids.

Data availability.

Next-generation sequencing data that support the findings of the study are available in the Gene Expression Omnibus using accession code GSE159786.

Code availability

The code that supports the findings of this study is available from the corresponding authors upon reasonable request.

References

  1. Davis, R. L., Weintraub, H. & Lassar, A. B. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell 51, 987–1000 (1987).

    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. Zhang, Y. et al. Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron 78, 785–798 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Parekh, U. et al. Mapping cellular reprogramming via pooled overexpression screens with paired fitness and single-cell RNA-sequencing readout. Cell Syst. 7, 548–555 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Tsunemoto, R. et al. Diverse reprogramming codes for neuronal identity. Nature 557, 375–380 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Pritsker, M., Ford, N. R., Jenq, H. T. & Lemischka, I. R. Genomewide gain-of-function genetic screen identifies functionally active genes in mouse embryonic stem cells. Proc. Natl Acad. Sci. USA 103, 6946–6951 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Theodorou, E. et al. A high throughput embryonic stem cell screen identifies Oct-2 as a bifunctional regulator of neuronal differentiation. Genes Dev. 23, 575–588 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yamamizu, K. et al. Identification of transcription factors for lineage-specific ESC differentiation. Stem Cell Rep. 1, 545–559 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. 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  Google Scholar 

  12. Lambert, S. A. et al. The human transcription factors. Cell 175, 598–599 (2018).

    Article  CAS  PubMed  Google Scholar 

  13. Nakatake, Y. et al. Generation and profiling of 2,135 human ESC lines for the systematic analyses of cell states perturbed by inducing single transcription factors. Cell Rep. 31, 107655 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Vaquerizas, J. M., Kummerfeld, S. K., Teichmann, S. A. & Luscombe, N. M. A census of human transcription factors: function, expression and evolution. Nat. Rev. Genet. 10, 252–263 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. Jolma, A. et al. DNA-binding specificities of human transcription factors. Cell 152, 327–339 (2013).

    Article  CAS  PubMed  Google Scholar 

  16. Seiler, C. Y. et al. DNASU plasmid and PSI:Biology-Materials repositories: resources to accelerate biological research. Nucleic Acids Res. 42, D1253–D1260 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Yang, X. et al. A public genome-scale lentiviral expression library of human ORFs. Nat. Methods 8, 659–661 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wiemann, S. et al. The ORFeome Collaboration: a genome-scale human ORF-clone resource. Nat. Methods 13, 191–192 (2016).

    Article  Google Scholar 

  19. Adewumi, O. et al. Characterization of human embryonic stem cell lines by the International Stem Cell Initiative. Nat. Biotechnol. 25, 803–816 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Busskamp, V. et al. Rapid neurogenesis through transcriptional activation in human stem cells. Mol. Syst. Biol. 10, 760 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Choi, J. et al. A comparison of genetically matched cell lines reveals the equivalence of human iPSCs and ESCs. Nat. Biotechnol. 33, 1173–1181 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Cahan, P. & Daley, G. Q. Origins and implications of pluripotent stem cell variability and heterogeneity. Nat. Rev. Mol. Cell Biol. 14, 357–368 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. Bermingham, N. A. et al. Math1: an essential gene for the generation of inner ear hair cells. Science 284, 1837–1841 (1999).

    Article  CAS  PubMed  Google Scholar 

  25. Sagal, J. et al. Proneural transcription factor Atoh1 drives highly efficient differentiation of human pluripotent stem cells into dopaminergic neurons. Stem Cells Transl. Med. 3, 888–898 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Xue, Y. et al. Synthetic mRNAs drive highly efficient iPS cell differentiation to dopaminergic neurons. Stem Cells Transl. Med. 8, 112–123 (2019).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Mai, T. et al. NKX3-1 is required for induced pluripotent stem cell reprogramming and can replace OCT4 in mouse and human iPSC induction. Nat. Cell Biol. 20, 900–908 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Radley, A. H. et al. Assessment of engineered cells using CellNet and RNA-seq. Nat. Protoc. 12, 1089–1102 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Liang, C. C., Park, A. Y. & Guan, J. L. In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro. Nat. Protoc. 2, 329–333 (2007).

    Article  CAS  PubMed  Google Scholar 

  31. Bell, E., Ivarsson, B. & Merrill, C. Production of a tissue-like structure by contraction of collagen lattices by human fibroblasts of different proliferative potential in vitro. Proc. Natl Acad. Sci. USA 76, 1274–1278 (1979).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Lee, D. et al. ER71 acts downstream of BMP, Notch, and Wnt signaling in blood and vessel progenitor specification. Cell Stem Cell 2, 497–507 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Baralle, F. E. & Giudice, J. Alternative splicing as a regulator of development and tissue identity. Nat. Rev. Mol. Cell Biol. 18, 437–451 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Potter, R. F. & Groom, A. C. Capillary diameter and geometry in cardiac and skeletal muscle studied by means of corrosion casts. Microvasc. Res. 25, 68–84 (1983).

    Article  CAS  PubMed  Google Scholar 

  35. Schaum, N. et al. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018).

    Article  PubMed Central  Google Scholar 

  36. Madhavan, M. et al. Induction of myelinating oligodendrocytes in human cortical spheroids. Nat. Methods 15, 700–706 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Marton, R. M. et al. Differentiation and maturation of oligodendrocytes in human three-dimensional neural cultures. Nat. Neurosci. 22, 484–491 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Garcia-Leon, J. A. et al. SOX10 single transcription factor-based fast and efficient generation of oligodendrocytes from human pluripotent stem cells. Stem Cell Rep. 10, 655–672 (2018).

    Article  CAS  Google Scholar 

  39. Ehrlich, M. et al. Rapid and efficient generation of oligodendrocytes from human induced pluripotent stem cells using transcription factors. Proc. Natl Acad. Sci. USA 114, E2243–E2252 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Sarkar, A. & Hochedlinger, K. The sox family of transcription factors: versatile regulators of stem and progenitor cell fate. Cell Stem Cell 12, 15–30 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Bi, W., Deng, J. M., Zhang, Z., Behringer, R. R. & de Crombrugghe, B. Sox9 is required for cartilage formation. Nat. Genet. 22, 85–89 (1999).

    Article  CAS  PubMed  Google Scholar 

  42. Canals, I. et al. Rapid and efficient induction of functional astrocytes from human pluripotent stem cells. Nat. Methods 15, 693–696 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Khoshakhlagh, P., Sivakumar, A., Pace, L. A., Sazer, D. W. & Moore, M. J. Methods for fabrication and evaluation of a 3D microengineered model of myelinated peripheral nerve. J. Neural Eng. 15, 064001 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Khoshakhlagh, P. & Moore, M. J. Photoreactive interpenetrating network of hyaluronic acid and Puramatrix as a selectively tunable scaffold for neurite growth. Acta Biomater. 16, 23–34 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Mohammadi, S. et al. Whole-brain in-vivo measurements of the axonal G-ratio in a group of 37 healthy volunteers. Front. Neurosci. 9, 441 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Windrem, M. S. et al. Fetal and adult human oligodendrocyte progenitor cell isolates myelinate the congenitally dysmyelinated brain. Nat. Med. 10, 93–97 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Lancaster, M. A. et al. Cerebral organoids model human brain development and microcephaly. Nature 501, 373–379 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Togo, S. et al. Differentiation of embryonic stem cells into fibroblast-like cells in three-dimensional type I collagen gel cultures. In Vitro Cell. Dev. Biol. Anim. 47, 114–124 (2011).

    Article  CAS  PubMed  Google Scholar 

  49. Elcheva, I. et al. Direct induction of haematoendothelial programs in human pluripotent stem cells by transcriptional regulators. Nat. Commun. 5, 4372 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Morita, R. et al. ETS transcription factor ETV2 directly converts human fibroblasts into functional endothelial cells. Proc. Natl Acad. Sci. USA 112, 160–165 (2015).

    Article  CAS  PubMed  Google Scholar 

  51. Cakir, B. et al. Engineering of human brain organoids with a functional vascular-like system. Nat. Methods 16, 1169–1175 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Woltjen, K. et al. piggyBac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature 458, 766–770 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ronaldson-Bouchard, K. & Vunjak-Novakovic, G. Organs-on-a-Chip: a fast track for engineered human tissues in drug development. Cell Stem Cell 22, 310–324 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Guye, P. et al. Genetically engineering self-organization of human pluripotent stem cells into a liver bud-like tissue using Gata6. Nat. Commun. 7, 10243 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Bagley, J. A., Reumann, D., Bian, S., Levi-Strauss, J. & Knoblich, J. A. Fused cerebral organoids model interactions between brain regions. Nat. Methods 14, 743–751 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Birey, F. et al. Assembly of functionally integrated human forebrain spheroids. Nature 545, 54–59 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Xiang, Y. et al. Fusion of regionally specified hPSC-derived organoids models human brain development and interneuron migration. Cell Stem Cell 21, 383–398 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Cederquist, G. Y. et al. Specification of positional identity in forebrain organoids. Nat. Biotechnol. 37, 436–444 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Mansour, A. A. et al. An in vivo model of functional and vascularized human brain organoids. Nat. Biotechnol. 36, 432–441 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. 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 

  61. Han, X. et al. Construction of a human cell landscape at single-cell level. Nature 581, 303–309 (2020).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Moss, J. et al. Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat. Commun. 9, 5068 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Gray, K. A., Yates, B., Seal, R. L., Wright, M. W. & Bruford, E. A. Genenames.org: the HGNC resources in 2015. Nucleic Acids Res. 43, D1079–D1085 (2015).

    Article  CAS  PubMed  Google Scholar 

  65. Mele, M. et al. Human genomics. The human transcriptome across tissues and individuals. Science 348, 660–665 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Church, G. M. The personal genome project. Mol. Syst. Biol. 1, 2005.0030 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kutsche, L. K. et al. Combined experimental and system-level analyses reveal the complex regulatory network of miR-124 during human neurogenesis. Cell Syst. 7, 438–452 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Salmon, P. & Trono, D. Production and titration of lentiviral vectors. in Current Protocols in Human Genetics Ch. 12, Unit 12.10 (Wiley, 2007).

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

    Article  CAS  PubMed  Google Scholar 

  70. 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 

  71. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Zhang, J. et al. A genome-wide analysis of human pluripotent stem cell-derived endothelial cells in 2D or 3D culture. Stem Cell Rep. 8, 907–918 (2017).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  74. Bult, C. J., Blake, J. A., Smith, C. L., Kadin, J. A. & Richardson, J. E. Mouse genome database (MGD) 2019. Nucleic Acids Res. 47, D801–D806 (2019).

    Article  CAS  PubMed  Google Scholar 

  75. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Leek, J. T. svaseq: removing batch effects and other unwanted noise from sequencing data. Nucleic Acids Res. 42, e161 (2014).

  77. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  PubMed  Google Scholar 

  78. Ngo, P., Ramalingam, P., Phillips, J. A. & Furuta, G. T. Collagen gel contraction assay. Methods Mol. Biol. 341, 103–109 (2006).

    CAS  PubMed  Google Scholar 

  79. Picelli, S. et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1098 (2013).

    Article  CAS  PubMed  Google Scholar 

  80. 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 

  81. 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 

  82. Koike, N. et al. Tissue engineering: creation of long-lasting blood vessels. Nature 428, 138–139 (2004).

    Article  CAS  PubMed  Google Scholar 

  83. Melero-Martin, J. M. et al. Engineering robust and functional vascular networks in vivo with human adult and cord blood-derived progenitor cells. Circ. Res. 103, 194–202 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Khoshakhlagh, P. et al. Development and characterization of a bioglass/chitosan composite as an injectable bone substitute. Carbohydrate Polym. 157, 1261–1271 (2017).

    Article  CAS  Google Scholar 

  85. Khoshakhlagh, P., Bowser, D. A., Brown, J. Q. & Moore, M. J. Comparison of visible and UVA phototoxicity in neural culture systems micropatterned with digital projection photolithography. J. Biomed. Mater. Res. A 107, 134–144 (2019).

    Article  CAS  PubMed  Google Scholar 

  86. Douvaras, P. & Fossati, V. Generation and isolation of oligodendrocyte progenitor cells from human pluripotent stem cells. Nat. Protoc. 10, 1143–1154 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank J. Aach, M. O. Karl, R. Kalhor, N. Ostrov and H. Lee for critical feedback and the Church and Busskamp laboratories for support. We acknowledge technical support from the Harvard Biopolymers Facility, the Harvard Division of Immunology Flow Cytometry Core Facility, the Beth Israel Deaconess Medical Center Flow Cytometry Core, the Wyss Flow Cytometry and Microscopy Core, M. Ericsson and P. Coughlin at the Harvard Medical School Electron Microscopy Facility, M. T. Gianatasio at the Dana-Farber/Harvard Cancer Center Specialized Histopathology Core and Rodent Histopathology Core (both supported, in part, by National Cancer Institute Cancer Center Support grant NIH 5 P30 CA06516) and Harvard Medical School Orchestra Research Computing. We also thank the TU Dresden Center for Molecular and Cellular Bioengineering Advanced Imaging, Deep Sequencing, Flow Cytometry and Stem Cell Engineering core facilities. We would also like to thank J. Gray’s laboratory for electrophysiology support, S. Jeanty and J. Lee (Church lab, Harvard Medical School) for the PGP1 Sendai virus hiPSC line, G. Sheynkman and W. Glindmeyer for helpful discussions, A. Jolma, K. Nitta and K. Said for technical assistance and M. Lemieux and J. McDade for their support in depositing the library to Addgene. A.H.M.N. was supported by an NSERC Postgraduate Fellowship and a Peter and Carolyn Lynch Foundation Fellowship. J.E.R.A. was supported by the DIGS-BB program. S.L.S. is a Shurl and Kay Curci Foundation Fellow of the Life Sciences Research Foundation. The Ellison Foundation and Institute Sponsored Research funds from the DFCI Strategic Initiative supported M.V. and D.E.H. The project was supported by the Volkswagen Foundation (Freigeist - A110720), the European Research Council (ERC-StG-678071 - ProNeurons) and the Deutsche Forschungsgemeinschaft (SPP2127, EXC-2068-390729961 - Cluster of Excellence - Physics of Life at TU Dresden and EXC-2151-390873048 – Cluster of Excellence – ImmunoSensation2 at the University of Bonn) to V.B. G.M.C. acknowledges funding from National Human Genome Research Institute grants P50 HG005550 ‘Center for Casual Variation’, RM1 HG008525 ‘Center for Genomically Engineered Organs’, the Simons Foundation for Autism Research Initiative (368485), the Blavatnik Biomedical Accelerator at Harvard University, the FunGCAT program from the Office of the Director of National Intelligence Intelligence Advanced Research Projects Activity, via the Army Research Office, under federal award no. W911NF-17-2-0089 and research funding from R. Merkin and the Merkin Family Foundation.

Author information

Authors and Affiliations

Authors

Contributions

A.H.M.N., P.K., V.B. and G.M.C. conceived the idea, led the study and designed all experiments. A.H.M.N. and P.K. performed most of the experiments and analyses, with significant technical contributions from J.E.R.A, G.P., K.W., A.S., S.L.S., E.A., K.K., R.E.K., A.V., M.D., K.L., W.S., J.Y.H., A.G., J.T., D.E.H., M.V. and J.M.M.-M. V.B. and G.M.C. oversaw the study. A.H.M.N., P.K. and V.B. wrote the manuscript with input and feedback from all authors.

Corresponding authors

Correspondence to Volker Busskamp or George M. Church.

Ethics declarations

Competing interests

A.H.M.N., P.K., V.B. and G.M.C. are inventors on patents filed by the Presidents and Fellows of Harvard College. Full disclosure for G.M.C. is available at http://arep.med.harvard.edu/gmc/tech.html. A.H.M.N., P.K. and G.M.C. are co-founders of and have equity in GC Therapeutics, Inc. No reagents or funding from GC Therapeutics were used in this study.

Additional information

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

Supplementary information

Supplementary Information

Supplementary Figs. 1–7

Reporting Summary

Supplementary Table 1

TFs in the Human TFome

Supplementary Table 2

TFome screen sequencing statistics

Supplementary Table 3

TFome screen differentiation scores

Supplementary Table 4

Novelty and tissue expression of 290 TF hits

Supplementary Table 5

RNA-seq statistics and expression profiles

Supplementary Table 6

TFs involved in oligodendrocyte development

Supplementary Table 7

Exact P values for statistical tests

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ng, A.H.M., Khoshakhlagh, P., Rojo Arias, J.E. et al. A comprehensive library of human transcription factors for cell fate engineering. Nat Biotechnol 39, 510–519 (2021). https://doi.org/10.1038/s41587-020-0742-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-020-0742-6

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research