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 lineage conversions: unnatural but useful?

Abstract

Classic experiments such as somatic cell nuclear transfer into oocytes and cell fusion demonstrated that differentiated cells are not irreversibly committed to their fate. More recent work has built on these conclusions and discovered defined factors that directly induce one specific cell type from another, which may be as distantly related as cells from different germ layers. This suggests the possibility that any specific cell type may be directly converted into any other if the appropriate reprogramming factors are known. Direct lineage conversion could provide important new sources of human cells for modeling disease processes or for cellular-replacement therapies. For future applications, it will be critical to carefully determine the fidelity of reprogramming and to develop methods for robustly and efficiently generating human cell types of interest.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Epigenetic models of development and reprogramming.
Figure 2: Various modes of induced cell fate changes.
Figure 3: Potential mechanism of the stepwise activation of silent genes by reprogramming factors.

Similar content being viewed by others

References

  1. Briggs, R. & King, T.J. Transplantation of living nuclei from blastula cells into enucleated frogs' eggs. Proc. Natl. Acad. Sci. USA 38, 455–463 (1952).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Gurdon, J.B., Elsdale, T.R. & Fischberg, M. Sexually mature individuals of Xenopus laevis from the transplantation of single somatic nuclei. Nature 182, 64–65 (1958).

    CAS  PubMed  Google Scholar 

  3. Gurdon, J.B. From nuclear transfer to nuclear reprogramming: the reversal of cell differentiation. Annu. Rev. Cell Dev. Biol. 22, 1–22 (2006).

    CAS  PubMed  Google Scholar 

  4. Gurdon, J.B. & Melton, D.A. Nuclear reprogramming in cells. Science 322, 1811–1815 (2008).

    CAS  PubMed  Google Scholar 

  5. Campbell, K.H., McWhir, J., Ritchie, W.A. & Wilmut, I. Sheep cloned by nuclear transfer from a cultured cell line. Nature 380, 64–66 (1996).

    CAS  PubMed  Google Scholar 

  6. Wilmut, I., Schnieke, A.E., McWhir, J., Kind, A.J. & Campbell, K.H. Viable offspring derived from fetal and adult mammalian cells. Nature 385, 810–813 (1997).

    CAS  PubMed  Google Scholar 

  7. Hochedlinger, K. & Jaenisch, R. Monoclonal mice generated by nuclear transfer from mature B and T donor cells. Nature 415, 1035–1038 (2002).

    CAS  PubMed  Google Scholar 

  8. Eggan, K. et al. Mice cloned from olfactory sensory neurons. Nature 428, 44–49 (2004).

    CAS  PubMed  Google Scholar 

  9. Li, J., Ishii, T., Feinstein, P. & Mombaerts, P. Odorant receptor gene choice is reset by nuclear transfer from mouse olfactory sensory neurons. Nature 428, 393–399 (2004).

    CAS  PubMed  Google Scholar 

  10. Bernstein, B.E., Meissner, A. & Lander, E.S. The mammalian epigenome. Cell 128, 669–681 (2007).

    CAS  PubMed  Google Scholar 

  11. Jenuwein, T. & Allis, C.D. Translating the histone code. Science 293, 1074–1080 (2001).

    CAS  PubMed  Google Scholar 

  12. Miller, R.A. & Ruddle, F.H. Pluripotent teratocarcinoma-thymus somatic cell hybrids. Cell 9, 45–55 (1976).

    CAS  PubMed  Google Scholar 

  13. Blau, H.M., Chiu, C.P. & Webster, C. Cytoplasmic activation of human nuclear genes in stable heterocaryons. Cell 32, 1171–1180 (1983).

    CAS  PubMed  Google Scholar 

  14. Blau, H.M. et al. Plasticity of the differentiated state. Science 230, 758–766 (1985).

    CAS  PubMed  Google Scholar 

  15. Tada, M., Takahama, Y., Abe, K., Nakatsuji, N. & Tada, T. Nuclear reprogramming of somatic cells by in vitro hybridization with ES cells. Curr. Biol. 11, 1553–1558 (2001).

    CAS  PubMed  Google Scholar 

  16. Cowan, C.A., Atienza, J., Melton, D.A. & Eggan, K. Nuclear reprogramming of somatic cells after fusion with human embryonic stem cells. Science 309, 1369–1373 (2005).

    CAS  PubMed  Google Scholar 

  17. Yamanaka, S. & Blau, H.M. Nuclear reprogramming to a pluripotent state by three approaches. Nature 465, 704–712 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. 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 

  19. Okita, K., Ichisaka, T. & Yamanaka, S. Generation of germline-competent induced pluripotent stem cells. Nature 448, 313–317 (2007).

    CAS  PubMed  Google Scholar 

  20. Wernig, M. et al. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448, 318–324 (2007).

    Article  CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  22. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    CAS  PubMed  Google Scholar 

  23. Park, I.H. et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature 451, 141–146 (2008).

    CAS  PubMed  Google Scholar 

  24. Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917–1920 (2007).

    CAS  PubMed  Google Scholar 

  25. Hochedlinger, K. & Plath, K. Epigenetic reprogramming and induced pluripotency. Development 136, 509–523 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Jaenisch, R. & Young, R. Stem cells, the molecular circuitry of pluripotency and nuclear reprogramming. Cell 132, 567–582 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Silva, J. & Smith, A. Capturing pluripotency. Cell 132, 532–536 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Pang, Z.P. et al. Induction of human neuronal cells by defined transcription factors. Nature 476, 220–223 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  32. Hochedlinger, K. & Jaenisch, R. Nuclear reprogramming and pluripotency. Nature 441, 1061–1067 (2006).

    CAS  PubMed  Google Scholar 

  33. Zhou, Q. & Melton, D.A. Extreme makeover: converting one cell into another. Cell Stem Cell 3, 382–388 (2008).

    CAS  PubMed  Google Scholar 

  34. Graf, T. & Enver, T. Forcing cells to change lineages. Nature 462, 587–594 (2009).

    CAS  PubMed  Google Scholar 

  35. Slack, J.M. Metaplasia and transdifferentiation: from pure biology to the clinic. Nat. Rev. Mol. Cell Biol. 8, 369–378 (2007).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Feng, R. et al. PU.1 and C/EBPalpha/beta convert fibroblasts into macrophage-like cells. Proc. Natl. Acad. Sci. USA 105, 6057–6062 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  39. Heinrich, C. et al. Directing astroglia from the cerebral cortex into subtype specific functional neurons. PLoS Biol. 8, e1000373 (2010).

    PubMed  PubMed Central  Google Scholar 

  40. Heins, N. et al. Glial cells generate neurons: the role of the transcription factor Pax6. Nat. Neurosci. 5, 308–315 (2002).

    CAS  PubMed  Google Scholar 

  41. Takeuchi, J.K. & Bruneau, B.G. Directed transdifferentiation of mouse mesoderm to heart tissue by defined factors. Nature 459, 708–711 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Ieda, M. et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell 142, 375–386 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Szabo, E. et al. Direct conversion of human fibroblasts to multilineage blood progenitors. Nature 468, 521–526 (2010).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  45. Taylor, S.M. & Jones, P.A. Multiple new phenotypes induced in 10T1/2 and 3T3 cells treated with 5-azacytidine. Cell 17, 771–779 (1979).

    CAS  PubMed  Google Scholar 

  46. Meissner, A. et al. Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature 454, 766–770 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Taberlay, P.C. & Jones, P.A. DNA methylation and cancer. Prog. Drug Res. 67, 1–23 (2011).

    CAS  PubMed  Google Scholar 

  48. De Carvalho, D.D., You, J.S. & Jones, P.A. DNA methylation and cellular reprogramming. Trends Cell Biol. 20, 609–617 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Chiu, C.P. & Blau, H.M. 5-Azacytidine permits gene activation in a previously noninducible cell type. Cell 40, 417–424 (1985).

    CAS  PubMed  Google Scholar 

  50. Weintraub, H. et al. Activation of muscle-specific genes in pigment, nerve, fat, liver, and fibroblast cell lines by forced expression of MyoD. Proc. Natl. Acad. Sci. USA 86, 5434–5438 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Choi, J. et al. MyoD converts primary dermal fibroblasts, chondroblasts, smooth muscle, and retinal pigmented epithelial cells into striated mononucleated myoblasts and multinucleated myotubes. Proc. Natl. Acad. Sci. USA 87, 7988–7992 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Lassar, A.B., Paterson, B.M. & Weintraub, H. Transfection of a DNA locus that mediates the conversion of 10T1/2 fibroblasts to myoblasts. Cell 47, 649–656 (1986).

    CAS  PubMed  Google Scholar 

  53. Weintraub, H. The MyoD family and myogenesis: redundancy, networks, and thresholds. Cell 75, 1241–1244 (1993).

    CAS  PubMed  Google Scholar 

  54. Schafer, B.W., Blakely, B.T., Darlington, G.J. & Blau, H.M. Effect of cell history on response to helix-loop-helix family of myogenic regulators. Nature 344, 454–458 (1990).

    CAS  PubMed  Google Scholar 

  55. Wright, W.E. Induction of muscle genes in neural cells. J. Cell Biol. 98, 427–435 (1984).

    CAS  PubMed  Google Scholar 

  56. Pavlath, G.K., Rich, K., Webster, S.G. & Blau, H.M. Localization of muscle gene products in nuclear domains. Nature 337, 570–573 (1989).

    CAS  PubMed  Google Scholar 

  57. Weintraub, H. et al. The myoD gene family: nodal point during specification of the muscle cell lineage. Science 251, 761–766 (1991).

    CAS  PubMed  Google Scholar 

  58. Blau, H.M. & Blakely, B.T. Plasticity of cell fate: insights from heterokaryons. Semin. Cell Dev. Biol. 10, 267–272 (1999).

    CAS  PubMed  Google Scholar 

  59. Pomerantz, J.H., Mukherjee, S., Palermo, A.T. & Blau, H.M. Reprogramming to a muscle fate by fusion recapitulates differentiation. J. Cell Sci. 122, 1045–1053 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Buckingham, M. et al. The formation of skeletal muscle: from somite to limb. J. Anat. 202, 59–68 (2003).

    PubMed  PubMed Central  Google Scholar 

  61. Kassar-Duchossoy, L. et al. Mrf4 determines skeletal muscle identity in Myf5:Myod double-mutant mice. Nature 431, 466–471 (2004).

    CAS  PubMed  Google Scholar 

  62. Hasty, P. et al. Muscle deficiency and neonatal death in mice with a targeted mutation in the myogenin gene. Nature 364, 501–506 (1993).

    CAS  PubMed  Google Scholar 

  63. Nabeshima, Y. et al. Myogenin gene disruption results in perinatal lethality because of severe muscle defect. Nature 364, 532–535 (1993).

    CAS  PubMed  Google Scholar 

  64. Berkes, C.A. & Tapscott, S.J. MyoD and the transcriptional control of myogenesis. Semin. Cell Dev. Biol. 16, 585–595 (2005).

    CAS  PubMed  Google Scholar 

  65. Tapscott, S.J. The circuitry of a master switch: MyoD and the regulation of skeletal muscle gene transcription. Development 132, 2685–2695 (2005).

    CAS  PubMed  Google Scholar 

  66. Blais, A. et al. An initial blueprint for myogenic differentiation. Genes Dev. 19, 553–569 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Berkes, C.A. et al. Pbx marks genes for activation by MyoD indicating a role for a homeodomain protein in establishing myogenic potential. Mol. Cell 14, 465–477 (2004).

    CAS  PubMed  Google Scholar 

  68. Albini, S. & Puri, P.L. SWI/SNF complexes, chromatin remodeling and skeletal myogenesis: it's time to exchange!. Exp. Cell Res. 316, 3073–3080 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Gerber, A.N., Klesert, T.R., Bergstrom, D.A. & Tapscott, S.J. Two domains of MyoD mediate transcriptional activation of genes in repressive chromatin: a mechanism for lineage determination in myogenesis. Genes Dev. 11, 436–450 (1997).

    CAS  PubMed  Google Scholar 

  70. Cao, Y. et al. Genome-wide MyoD binding in skeletal muscle cells: a potential for broad cellular reprogramming. Dev. Cell 18, 662–674 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Molkentin, J.D., Black, B.L., Martin, J.F. & Olson, E.N. Cooperative activation of muscle gene expression by MEF2 and myogenic bHLH proteins. Cell 83, 1125–1136 (1995).

    CAS  PubMed  Google Scholar 

  72. Bergstrom, D.A. et al. Promoter-specific regulation of MyoD binding and signal transduction cooperate to pattern gene expression. Mol. Cell 9, 587–600 (2002).

    CAS  PubMed  Google Scholar 

  73. de la Serna, I.L. et al. MyoD targets chromatin remodeling complexes to the myogenin locus prior to forming a stable DNA-bound complex. Mol. Cell Biol. 25, 3997–4009 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Puri, P.L. et al. Differential roles of p300 and PCAF acetyltransferases in muscle differentiation. Mol. Cell 1, 35–45 (1997).

    CAS  PubMed  Google Scholar 

  75. Seale, P. et al. PRDM16 controls a brown fat/skeletal muscle switch. Nature 454, 961–967 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Kajimura, S., Seale, P. & Spiegelman, B.M. Transcriptional control of brown fat development. Cell Metab. 11, 257–262 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Kajimura, S. et al. Initiation of myoblast to brown fat switch by a PRDM16-C/EBP-beta transcriptional complex. Nature 460, 1154–1158 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Olson, E.N. Gene regulatory networks in the evolution and development of the heart. Science 313, 1922–1927 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Srivastava, D. Making or breaking the heart: from lineage determination to morphogenesis. Cell 126, 1037–1048 (2006).

    CAS  PubMed  Google Scholar 

  80. Lickert, H. et al. Baf60c is essential for function of BAF chromatin remodelling complexes in heart development. Nature 432, 107–112 (2004).

    CAS  PubMed  Google Scholar 

  81. 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).

    CAS  PubMed  Google Scholar 

  82. Smale, S.T. Pioneer factors in embryonic stem cells and differentiation. Curr. Opin. Genet. Dev. 20, 519–526 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Davidson, E.H. & Levine, M.S. Properties of developmental gene regulatory networks. Proc. Natl. Acad. Sci. USA 105, 20063–20066 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Reya, T., Morrison, S.J., Clarke, M.F. & Weissman, I.L. Stem cells, cancer, and cancer stem cells. Nature 414, 105–111 (2001).

    CAS  PubMed  Google Scholar 

  85. Orkin, S.H. & Zon, L.I. Hematopoiesis: an evolving paradigm for stem cell biology. Cell 132, 631–644 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Rovigatti, U. et al. Heavy chain immunoglobulin gene rearrangement in acute nonlymphocytic leukemia. Blood 63, 1023–1027 (1984).

    CAS  PubMed  Google Scholar 

  87. Ha, K., Minden, M., Hozumi, N. & Gelfand, E.W. Immunoglobulin gene rearrangement in acute myelogenous leukemia. Cancer Res. 44, 4658–4660 (1984).

    CAS  PubMed  Google Scholar 

  88. Palumbo, A., Minowada, J., Erikson, J., Croce, C.M. & Rovera, G. Lineage infidelity of a human myelogenous leukemia cell line. Blood 64, 1059–1063 (1984).

    CAS  PubMed  Google Scholar 

  89. Tsai, S.F. et al. Cloning of cDNA for the major DNA-binding protein of the erythroid lineage through expression in mammalian cells. Nature 339, 446–451 (1989).

    CAS  PubMed  Google Scholar 

  90. Wall, L., deBoer, E. & Grosveld, F. The human beta-globin gene 3′ enhancer contains multiple binding sites for an erythroid-specific protein. Genes Dev. 2, 1089–1100 (1988).

    CAS  PubMed  Google Scholar 

  91. Evans, T., Reitman, M. & Felsenfeld, G. An erythrocyte-specific DNA-binding factor recognizes a regulatory sequence common to all chicken globin genes. Proc. Natl. Acad. Sci. USA 85, 5976–5980 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Pevny, L. et al. Erythroid differentiation in chimaeric mice blocked by a targeted mutation in the gene for transcription factor GATA-1. Nature 349, 257–260 (1991).

    CAS  PubMed  Google Scholar 

  93. Visvader, J.E., Elefanty, A.G., Strasser, A. & Adams, J.M. GATA-1 but not SCL induces megakaryocytic differentiation in an early myeloid line. EMBO J. 11, 4557–4564 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  95. Nerlov, C. & Graf, T. PU.1 induces myeloid lineage commitment in multipotent hematopoietic progenitors. Genes Dev. 12, 2403–2412 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Heyworth, C., Pearson, S., May, G. & Enver, T. Transcription factor-mediated lineage switching reveals plasticity in primary committed progenitor cells. EMBO J. 21, 3770–3781 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Kondo, M. et al. Cell-fate conversion of lymphoid-committed progenitors by instructive actions of cytokines. Nature 407, 383–386 (2000).

    CAS  PubMed  Google Scholar 

  98. Rolink, A.G., Nutt, S.L., Melchers, F. & Busslinger, M. Long-term in vivo reconstitution of T-cell development by Pax5-deficient B-cell progenitors. Nature 401, 603–606 (1999).

    CAS  PubMed  Google Scholar 

  99. Mikkola, I., Heavey, B., Horcher, M. & Busslinger, M. Reversion of B cell commitment upon loss of Pax5 expression. Science 297, 110–113 (2002).

    CAS  PubMed  Google Scholar 

  100. Cobaleda, C., Jochum, W. & Busslinger, M. Conversion of mature B cells into T cells by dedifferentiation to uncommitted progenitors. Nature 449, 473–477 (2007).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Ghisletti, S. et al. Identification and characterization of enhancers controlling the inflammatory gene expression program in macrophages. Immunity 32, 317–328 (2010).

    CAS  PubMed  Google Scholar 

  103. Natoli, G. Maintaining cell identity through global control of genomic organization. Immunity 33, 12–24 (2010).

    CAS  PubMed  Google Scholar 

  104. Visel, A., Rubin, E.M. & Pennacchio, L.A. Genomic views of distant-acting enhancers. Nature 461, 199–205 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Visel, A. et al. ChIP-seq accurately predicts tissue-specific activity of enhancers. Nature 457, 854–858 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470, 279–283 (2011).

    CAS  PubMed  Google Scholar 

  107. Creyghton, M.P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl. Acad. Sci. USA 107, 21931–21936 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Bussmann, L.H. et al. A robust and highly efficient immune cell reprogramming system. Cell Stem Cell 5, 554–566 (2009).

    CAS  PubMed  Google Scholar 

  109. Hochedlinger, K., Yamada, Y., Beard, C. & Jaenisch, R. Ectopic expression of Oct-4 blocks progenitor-cell differentiation and causes dysplasia in epithelial tissues. Cell 121, 465–477 (2005).

    CAS  PubMed  Google Scholar 

  110. Lengner, C.J. et al. Oct4 expression is not required for mouse somatic stem cell self-renewal. Cell Stem Cell 1, 403–415 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Wang, V.E., Schmidt, T., Chen, J., Sharp, P.A. & Tantin, D. Embryonic lethality, decreased erythropoiesis, and defective octamer-dependent promoter activation in Oct-1-deficient mice. Mol. Cell. Biol. 24, 1022–1032 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Sebastiano, V. et al. Oct1 regulates trophoblast development during early mouse embryogenesis. Development 137, 3551–3560 (2010).

    CAS  PubMed  Google Scholar 

  113. Corcoran, L.M. et al. Oct-2, although not required for early B-cell development, is critical for later B-cell maturation and for postnatal survival. Genes Dev. 7, 570–582 (1993).

    CAS  PubMed  Google Scholar 

  114. Zhou, Q. et al. A multipotent progenitor domain guides pancreatic organogenesis. Dev. Cell 13, 103–114 (2007).

    CAS  PubMed  Google Scholar 

  115. Zaret, K.S. Genetic programming of liver and pancreas progenitors: lessons for stem-cell differentiation. Nat. Rev. Genet. 9, 329–340 (2008).

    CAS  PubMed  Google Scholar 

  116. Zaret, K.S. & Grompe, M. Generation and regeneration of cells of the liver and pancreas. Science 322, 1490–1494 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Murtaugh, L.C. & Melton, D.A. Genes, signals, and lineages in pancreas development. Annu. Rev. Cell Dev. Biol. 19, 71–89 (2003).

    CAS  PubMed  Google Scholar 

  118. Bonal, C. & Herrera, P.L. Genes controlling pancreas ontogeny. Int. J. Dev. Biol. 52, 823–835 (2008).

    CAS  PubMed  Google Scholar 

  119. Puri, S. & Hebrok, M. Cellular plasticity within the pancreas–lessons learned from development. Dev. Cell 18, 342–356 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Dor, Y., Brown, J., Martinez, O.I. & Melton, D.A. Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nature 429, 41–46 (2004).

    CAS  PubMed  Google Scholar 

  121. Teta, M., Rankin, M.M., Long, S.Y., Stein, G.M. & Kushner, J.A. Growth and regeneration of adult beta cells does not involve specialized progenitors. Dev. Cell 12, 817–826 (2007).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Zaret, K.S. & White, M.F. Diabetes forum: extreme makeover of pancreatic alpha-cells. Nature 464, 1132–1133 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Xu, X. et al. Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell 132, 197–207 (2008).

    CAS  PubMed  Google Scholar 

  125. Shen, C.N., Slack, J.M. & Tosh, D. Molecular basis of transdifferentiation of pancreas to liver. Nat. Cell Biol. 2, 879–887 (2000).

    CAS  PubMed  Google Scholar 

  126. Ferber, S. et al. Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia. Nat. Med. 6, 568–572 (2000).

    CAS  PubMed  Google Scholar 

  127. Kaneto, H. et al. PDX-1/VP16 fusion protein, together with NeuroD or Ngn3, markedly induces insulin gene transcription and ameliorates glucose tolerance. Diabetes 54, 1009–1022 (2005).

    CAS  PubMed  Google Scholar 

  128. Miyatsuka, T. et al. Ectopically expressed PDX-1 in liver initiates endocrine and exocrine pancreas differentiation but causes dysmorphogenesis. Biochem. Biophys. Res. Commun. 310, 1017–1025 (2003).

    CAS  PubMed  Google Scholar 

  129. Sapir, T. et al. Cell-replacement therapy for diabetes: generating functional insulin-producing tissue from adult human liver cells. Proc. Natl. Acad. Sci. USA 102, 7964–7969 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Horb, M.E., Shen, C.N., Tosh, D. & Slack, J.M. Experimental conversion of liver to pancreas. Curr. Biol. 13, 105–115 (2003).

    CAS  PubMed  Google Scholar 

  131. Grapin-Botton, A., Majithia, A.R. & Melton, D.A. Key events of pancreas formation are triggered in gut endoderm by ectopic expression of pancreatic regulatory genes. Genes Dev. 15, 444–454 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Gu, G., Dubauskaite, J. & Melton, D.A. Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors. Development 129, 2447–2457 (2002).

    CAS  PubMed  Google Scholar 

  133. Jonsson, J., Carlsson, L., Edlund, T. & Edlund, H. Insulin-promoter-factor 1 is required for pancreas development in mice. Nature 371, 606–609 (1994).

    CAS  PubMed  Google Scholar 

  134. Ahlgren, U., Jonsson, J., Jonsson, L., Simu, K. & Edlund, H. Beta-cell-specific inactivation of the mouse Ipf1/Pdx1 gene results in loss of the beta-cell phenotype and maturity onset diabetes. Genes Dev. 12, 1763–1768 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Waeber, G., Thompson, N., Nicod, P. & Bonny, C. Transcriptional activation of the GLUT2 gene by the IPF-1/STF-1/IDX-1 homeobox factor. Mol. Endocrinol. 10, 1327–1334 (1996).

    CAS  PubMed  Google Scholar 

  136. Watada, H. et al. The human glucokinase gene beta-cell-type promoter: an essential role of insulin promoter factor 1/PDX-1 in its activation in HIT-T15 cells. Diabetes 45, 1478–1488 (1996).

    CAS  PubMed  Google Scholar 

  137. Apelqvist, A. et al. Notch signalling controls pancreatic cell differentiation. Nature 400, 877–881 (1999).

    CAS  PubMed  Google Scholar 

  138. Yechoor, V. et al. Neurogenin3 is sufficient for transdetermination of hepatic progenitor cells into neo-islets in vivo but not transdifferentiation of hepatocytes. Dev. Cell 16, 358–373 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Desgraz, R. & Herrera, P.L. Pancreatic neurogenin 3-expressing cells are unipotent islet precursors. Development 136, 3567–3574 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Gradwohl, G., Dierich, A., LeMeur, M. & Guillemot, F. Neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas. Proc. Natl. Acad. Sci. USA 97, 1607–1611 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Wang, S. et al. Sustained Neurog3 expression in hormone-expressing islet cells is required for endocrine maturation and function. Proc. Natl. Acad. Sci. USA 106, 9715–9720 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Gasa, R. et al. Induction of pancreatic islet cell differentiation by the neurogenin-neuroD cascade. Differentiation 76, 381–391 (2008).

    CAS  PubMed  Google Scholar 

  143. Gasa, R. et al. Proendocrine genes coordinate the pancreatic islet differentiation program in vitro. Proc. Natl. Acad. Sci. USA 101, 13245–13250 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Gu, C. et al. Pancreatic beta cells require NeuroD to achieve and maintain functional maturity. Cell Metab. 11, 298–310 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Kojima, H. et al. NeuroD-betacellulin gene therapy induces islet neogenesis in the liver and reverses diabetes in mice. Nat. Med. 9, 596–603 (2003).

    CAS  PubMed  Google Scholar 

  146. Bertrand, N., Castro, D.S. & Guillemot, F. Proneural genes and the specification of neural cell types. Nat. Rev. Neurosci. 3, 517–530 (2002).

    CAS  PubMed  Google Scholar 

  147. Olbrot, M., Rud, J., Moss, L.G. & Sharma, A. Identification of beta-cell-specific insulin gene transcription factor RIPE3b1 as mammalian MafA. Proc. Natl. Acad. Sci. USA 99, 6737–6742 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Kataoka, K. et al. MafA is a glucose-regulated and pancreatic beta-cell-specific transcriptional activator for the insulin gene. J. Biol. Chem. 277, 49903–49910 (2002).

    CAS  PubMed  Google Scholar 

  149. Matsuoka, T.A. et al. Members of the large Maf transcription family regulate insulin gene transcription in islet beta cells. Mol. Cell Biol. 23, 6049–6062 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Artner, I., Hang, Y., Guo, M., Gu, G. & Stein, R. MafA is a dedicated activator of the insulin gene in vivo. J. Endocrinol. 198, 271–279 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Zhang, C. et al. MafA is a key regulator of glucose-stimulated insulin secretion. Mol. Cell Biol. 25, 4969–4976 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Nishimura, W., Bonner-Weir, S. & Sharma, A. Expression of MafA in pancreatic progenitors is detrimental for pancreatic development. Dev. Biol. 333, 108–120 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Matsuoka, T.A. et al. The MafA transcription factor appears to be responsible for tissue-specific expression of insulin. Proc. Natl. Acad. Sci. USA 101, 2930–2933 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Nishimura, W. et al. A switch from MafB to MafA expression accompanies differentiation to pancreatic beta-cells. Dev. Biol. 293, 526–539 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Kaneto, H. et al. A crucial role of MafA as a novel therapeutic target for diabetes. J. Biol. Chem. 280, 15047–15052 (2005).

    CAS  PubMed  Google Scholar 

  156. Wilson, S.W. & Houart, C. Early steps in the development of the forebrain. Dev. Cell 6, 167–181 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Caspary, T. & Anderson, K.V. Patterning cell types in the dorsal spinal cord: what the mouse mutants say. Nat. Rev. Neurosci. 4, 289–297 (2003).

    PubMed  Google Scholar 

  158. Jessell, T.M. Neuronal specification in the spinal cord: inductive signals and transcriptional codes. Nat. Rev. Genet. 1, 20–29 (2000).

    CAS  PubMed  Google Scholar 

  159. Guillemot, F. Cellular and molecular control of neurogenesis in the mammalian telencephalon. Curr. Opin. Cell Biol. 17, 639–647 (2005).

    CAS  PubMed  Google Scholar 

  160. Molyneaux, B.J., Arlotta, P., Menezes, J.R. & Macklis, J.D. Neuronal subtype specification in the cerebral cortex. Nat. Rev. Neurosci. 8, 427–437 (2007).

    CAS  PubMed  Google Scholar 

  161. Wonders, C.P. & Anderson, S.A. The origin and specification of cortical interneurons. Nat. Rev. Neurosci. 7, 687–696 (2006).

    CAS  PubMed  Google Scholar 

  162. Rowitch, D.H. Glial specification in the vertebrate neural tube. Nat. Rev. Neurosci. 5, 409–419 (2004).

    CAS  PubMed  Google Scholar 

  163. Jan, Y.N. & Jan, L.Y. Neuronal cell fate specification in Drosophila. Curr. Opin. Neurobiol. 4, 8–13 (1994).

    CAS  PubMed  Google Scholar 

  164. Ross, S.E., Greenberg, M.E. & Stiles, C.D. Basic helix-loop-helix factors in cortical development. Neuron 39, 13–25 (2003).

    CAS  PubMed  Google Scholar 

  165. Lee, J.E. et al. Conversion of Xenopus ectoderm into neurons by NeuroD, a basic helix-loop-helix protein. Science 268, 836–844 (1995).

    CAS  PubMed  Google Scholar 

  166. Turner, D.L. & Weintraub, H. Expression of achaete-scute homolog 3 in Xenopus embryos converts ectodermal cells to a neural fate. Genes Dev. 8, 1434–1447 (1994).

    CAS  PubMed  Google Scholar 

  167. Bellefroid, E.J. et al. X-MyT1, a Xenopus C2HC-type zinc finger protein with a regulatory function in neuronal differentiation. Cell 87, 1191–1202 (1996).

    CAS  PubMed  Google Scholar 

  168. Perez, S.E., Rebelo, S. & Anderson, D.J. Early specification of sensory neuron fate revealed by expression and function of neurogenins in the chick embryo. Development 126, 1715–1728 (1999).

    CAS  PubMed  Google Scholar 

  169. Ma, Q., Kintner, C. & Anderson, D.J. Identification of neurogenin, a vertebrate neuronal determination gene. Cell 87, 43–52 (1996).

    CAS  PubMed  Google Scholar 

  170. Farah, M.H. et al. Generation of neurons by transient expression of neural bHLH proteins in mammalian cells. Development 127, 693–702 (2000).

    CAS  PubMed  Google Scholar 

  171. Nieto, M., Schuurmans, C., Britz, O. & Guillemot, F. Neural bHLH genes control the neuronal versus glial fate decision in cortical progenitors. Neuron 29, 401–413 (2001).

    CAS  PubMed  Google Scholar 

  172. Lo, L., Sommer, L. & Anderson, D.J. MASH1 maintains competence for BMP2-induced neuronal differentiation in post-migratory neural crest cells. Curr. Biol. 7, 440–450 (1997).

    CAS  PubMed  Google Scholar 

  173. Louvi, A. & Artavanis-Tsakonas, S. Notch signalling in vertebrate neural development. Nat. Rev. Neurosci. 7, 93–102 (2006).

    CAS  PubMed  Google Scholar 

  174. Casarosa, S., Fode, C. & Guillemot, F. Mash1 regulates neurogenesis in the ventral telencephalon. Development 126, 525–534 (1999).

    CAS  PubMed  Google Scholar 

  175. Fode, C. et al. A role for neural determination genes in specifying the dorsoventral identity of telencephalic neurons. Genes Dev. 14, 67–80 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Parras, C.M. et al. Divergent functions of the proneural genes Mash1 and Ngn2 in the specification of neuronal subtype identity. Genes Dev. 16, 324–338 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Sommer, L., Shah, N., Rao, M. & Anderson, D.J. The cellular function of MASH1 in autonomic neurogenesis. Neuron 15, 1245–1258 (1995).

    CAS  PubMed  Google Scholar 

  178. Guillemot, F. Spatial and temporal specification of neural fates by transcription factor codes. Development 134, 3771–3780 (2007).

    CAS  PubMed  Google Scholar 

  179. Shirasaki, R. & Pfaff, S.L. Transcriptional codes and the control of neuronal identity. Annu. Rev. Neurosci. 25, 251–281 (2002).

    CAS  PubMed  Google Scholar 

  180. Guillemot, F. et al. Mammalian achaete-scute homolog 1 is required for the early development of olfactory and autonomic neurons. Cell 75, 463–476 (1993).

    CAS  PubMed  Google Scholar 

  181. Kondo, T. & Raff, M. Basic helix-loop-helix proteins and the timing of oligodendrocyte differentiation. Development 127, 2989–2998 (2000).

    CAS  PubMed  Google Scholar 

  182. Sugimori, M. et al. Ascl1 is required for oligodendrocyte development in the spinal cord. Development 135, 1271–1281 (2008).

    CAS  PubMed  Google Scholar 

  183. Parras, C.M. et al. The proneural gene Mash1 specifies an early population of telencephalic oligodendrocytes. J. Neurosci. 27, 4233–4242 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Petryniak, M.A., Potter, G.B., Rowitch, D.H. & Rubenstein, J.L. Dlx1 and Dlx2 control neuronal versus oligodendroglial cell fate acquisition in the developing forebrain. Neuron 55, 417–433 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Jessberger, S., Toni, N., Clemenson, G.D. Jr., Ray, J. & Gage, F.H. Directed differentiation of hippocampal stem/progenitor cells in the adult brain. Nat. Neurosci. 11, 888–893 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Kriegstein, A. & Alvarez-Buylla, A. The glial nature of embryonic and adult neural stem cells. Annu. Rev. Neurosci. 32, 149–184 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Kondo, T. & Raff, M. Oligodendrocyte precursor cells reprogrammed to become multipotential CNS stem cells. Science 289, 1754–1757 (2000).

    CAS  PubMed  Google Scholar 

  188. Malatesta, P., Hartfuss, E. & Gotz, M. Isolation of radial glial cells by fluorescent-activated cell sorting reveals a neuronal lineage. Development 127, 5253–5263 (2000).

    CAS  PubMed  Google Scholar 

  189. Noctor, S.C., Flint, A.C., Weissman, T.A., Dammerman, R.S. & Kriegstein, A.R. Neurons derived from radial glial cells establish radial units in neocortex. Nature 409, 714–720 (2001).

    CAS  PubMed  Google Scholar 

  190. Berninger, B. et al. Functional properties of neurons derived from in vitro reprogrammed postnatal astroglia. J. Neurosci. 27, 8654–8664 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Mori, T. et al. Inducible gene deletion in astroglia and radial glia–a valuable tool for functional and lineage analysis. Glia 54, 21–34 (2006).

    PubMed  Google Scholar 

  192. Blum, R. et al. Neuronal network formation from reprogrammed early postnatal rat cortical glial cells. Cereb. Cortex 21, 413–424 (2011).

    PubMed  Google Scholar 

  193. Gotz, M. Making glutamatergic neurons from GABAergic progenitors. Nat. Neurosci. 13, 1308–1309 (2010).

    PubMed  Google Scholar 

  194. Arber, S. et al. Requirement for the homeobox gene Hb9 in the consolidation of motor neuron identity. Neuron 23, 659–674 (1999).

    CAS  PubMed  Google Scholar 

  195. Rouaux, C. & Arlotta, P. Fezf2 directs the differentiation of corticofugal neurons from striatal progenitors in vivo. Nat. Neurosci. 13, 1345–1347 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Cheng, L. et al. Tlx3 and Tlx1 are post-mitotic selector genes determining glutamatergic over GABAergic cell fates. Nat. Neurosci. 7, 510–517 (2004).

    CAS  PubMed  Google Scholar 

  197. Leone, D.P., Srinivasan, K., Chen, B., Alcamo, E. & McConnell, S.K. The determination of projection neuron identity in the developing cerebral cortex. Curr. Opin. Neurobiol. 18, 28–35 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  198. McEvilly, R.J., de Diaz, M.O., Schonemann, M.D., Hooshmand, F. & Rosenfeld, M.G. Transcriptional regulation of cortical neuron migration by POU domain factors. Science 295, 1528–1532 (2002).

    CAS  PubMed  Google Scholar 

  199. Sugitani, Y. et al. Brn-1 and Brn-2 share crucial roles in the production and positioning of mouse neocortical neurons. Genes Dev. 16, 1760–1765 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Pfisterer, U. et al. Direct conversion of human fibroblasts to dopaminergic neurons. Proc. Natl. Acad. Sci. USA 108, 10343–10348 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Caiazzo, M. et al. Direct generation of functional dopaminergic neurons from mouse and human fibroblasts. Nature 476, 224–227 (2011).

    CAS  PubMed  Google Scholar 

  203. Wang, S. et al. Myt1 and Ngn3 form a feed-forward expression loop to promote endocrine islet cell differentiation. Dev. Biol. 317, 531–540 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  204. Weiner, J.A. & Chun, J. Png-1, a nervous system-specific zinc finger gene, identifies regions containing postmitotic neurons during mammalian embryonic development. J. Comp. Neurol. 381, 130–142 (1997).

    CAS  PubMed  Google Scholar 

  205. Cahoy, J.D. et al. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J. Neurosci. 28, 264–278 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Armstrong, R.C., Kim, J.G. & Hudson, L.D. Expression of myelin transcription factor I (MyTI), a “zinc-finger” DNA-binding protein, in developing oligodendrocytes. Glia 14, 303–321 (1995).

    CAS  PubMed  Google Scholar 

  207. Gu, G. et al. Global expression analysis of gene regulatory pathways during endocrine pancreatic development. Development 131, 165–179 (2004).

    CAS  PubMed  Google Scholar 

  208. Castro, D.S. et al. Proneural bHLH and Brn proteins coregulate a neurogenic program through cooperative binding to a conserved DNA motif. Dev. Cell 11, 831–844 (2006).

    CAS  PubMed  Google Scholar 

  209. Witta, S.E., Agarwal, V.R. & Sato, S.M. XIPOU 2, a noggin-inducible gene, has direct neuralizing activity. Development 121, 721–730 (1995).

    CAS  PubMed  Google Scholar 

  210. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  211. Tursun, B., Patel, T., Kratsios, P. & Hobert, O. Direct conversion of C. elegans germ cells into specific neuron types. Science 331, 304–308 (2011).

    CAS  PubMed  Google Scholar 

  212. Singhal, N. et al. Chromatin-remodeling components of the BAF complex facilitate reprogramming. Cell 141, 943–955 (2010).

    CAS  PubMed  Google Scholar 

  213. Anokye-Danso, F. et al. Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell 8, 376–388 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Miyoshi, N. et al. Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell 8, 633–638 (2011).

    CAS  PubMed  Google Scholar 

  215. Goldsmith, M.A., Desai, D.M., Schultz, T. & Weiss, A. Function of a heterologous muscarinic receptor in T cell antigen receptor signal transduction mutants. J. Biol. Chem. 264, 17190–17197 (1989).

    CAS  PubMed  Google Scholar 

  216. Fambrough, D., McClure, K., Kazlauskas, A. & Lander, E.S. Diverse signaling pathways activated by growth factor receptors induce broadly overlapping, rather than independent, sets of genes. Cell 97, 727–741 (1999).

    CAS  PubMed  Google Scholar 

  217. Davidson, E.H. Emerging properties of animal gene regulatory networks. Nature 468, 911–920 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Probst, A.V., Dunleavy, E. & Almouzni, G. Epigenetic inheritance during the cell cycle. Nat. Rev. Mol. Cell Biol. 10, 192–206 (2009).

    CAS  PubMed  Google Scholar 

  219. Hanna, J. et al. Direct cell reprogramming is a stochastic process amenable to acceleration. Nature 462, 595–601 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  220. Hong, H. et al. Suppression of induced pluripotent stem cell generation by the p53-p21 pathway. Nature 460, 1132–1135 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  221. Utikal, J. et al. Immortalization eliminates a roadblock during cellular reprogramming into iPS cells. Nature 460, 1145–1148 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  222. Marion, R.M. et al. A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature 460, 1149–1153 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  223. Li, H. et al. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature 460, 1136–1139 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  224. Kawamura, T. et al. Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature 460, 1140–1144 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  225. Bhutani, N. et al. Reprogramming towards pluripotency requires AID-dependent DNA demethylation. Nature 463, 1042–1047 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  226. Meissner, A., Wernig, M. & Jaenisch, R. Direct reprogramming of genetically unmodified fibroblasts into pluripotent stem cells. Nat. Biotechnol. 25, 1177–1181 (2007).

    CAS  PubMed  Google Scholar 

  227. Wernig, M. et al. A drug-inducible transgenic system for direct reprogramming of multiple somatic cell types. Nat. Biotechnol. 26, 916–924 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  228. Ptashne, M. & Gann, A. Genes & Signals (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 2002).

  229. Davidson, E.H. The Regulatory Genome: Gene Regulatory Networks in Development and Evolution (Academic, Burlington, MA; San Diego, 2006).

  230. Li, B., Carey, M. & Workman, J.L. The role of chromatin during transcription. Cell 128, 707–719 (2007).

    CAS  PubMed  Google Scholar 

  231. Ho, L. & Crabtree, G.R. Chromatin remodelling during development. Nature 463, 474–484 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  232. Suzuki, M.M. & Bird, A. DNA methylation landscapes: provocative insights from epigenomics. Nat. Rev. Genet. 9, 465–476 (2008).

    CAS  PubMed  Google Scholar 

  233. Ma, D.K. et al. Neuronal activity-induced Gadd45b promotes epigenetic DNA demethylation and adult neurogenesis. Science 323, 1074–1077 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  234. Zaret, K.S. et al. Pioneer factors, genetic competence, and inductive signaling: programming liver and pancreas progenitors from the endoderm. Cold Spring Harb. Symp. Quant. Biol. 73, 119–126 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  235. Niwa, H. How is pluripotency determined and maintained? Development 134, 635–646 (2007).

    CAS  PubMed  Google Scholar 

  236. Erwin, D.H. & Davidson, E.H. The evolution of hierarchical gene regulatory networks. Nat. Rev. Genet. 10, 141–148 (2009).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  238. Mikkelsen, T.S. et al. Dissecting direct reprogramming through integrative genomic analysis. Nature 454, 49–55 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  239. Aoi, T. et al. Generation of pluripotent stem cells from adult mouse liver and stomach cells. Science 321, 699–702 (2008).

    CAS  PubMed  Google Scholar 

  240. Hanna, J. et al. Direct reprogramming of terminally differentiated mature B lymphocytes to pluripotency. Cell 133, 250–264 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  241. Sun, Y. et al. Neurogenin promotes neurogenesis and inhibits glial differentiation by independent mechanisms. Cell 104, 365–376 (2001).

    CAS  PubMed  Google Scholar 

  242. Frankel, N. et al. Phenotypic robustness conferred by apparently redundant transcriptional enhancers. Nature 466, 490–493 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  243. Ng, R.K. & Gurdon, J.B. Epigenetic memory of active gene transcription is inherited through somatic cell nuclear transfer. Proc. Natl. Acad. Sci. USA 102, 1957–1962 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  244. Biddle, A., Simeoni, I. & Gurdon, J.B. Xenopus oocytes reactivate muscle gene transcription in transplanted somatic nuclei independently of myogenic factors. Development 136, 2695–2703 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  245. Polo, J.M. et al. Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nat. Biotechnol. 28, 848–855 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  246. Kim, K. et al. Epigenetic memory in induced pluripotent stem cells. Nature 467, 285–290 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  247. Gaspard, N., Gaillard, A. & Vanderhaeghen, P. Making cortex in a dish: in vitro corticopoiesis from embryonic stem cells. Cell Cycle 8, 2491–2496 (2009).

    CAS  PubMed  Google Scholar 

  248. Saha, K. & Jaenisch, R. Technical challenges in using human induced pluripotent stem cells to model disease. Cell Stem Cell 5, 584–595 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  249. Stadtfeld, M. & Hochedlinger, K. Induced pluripotency: history, mechanisms, and applications. Genes Dev. 24, 2239–2263 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  250. Lin, W. et al. Foxa1 and Foxa2 function both upstream of and cooperatively with Lmx1a and Lmx1b in a feedforward loop promoting mesodiencephalic dopaminergic neuron development. Dev. Biol. 333, 386–396 (2009).

    CAS  PubMed  Google Scholar 

  251. Omodei, D. et al. Anterior-posterior graded response to Otx2 controls proliferation and differentiation of dopaminergic progenitors in the ventral mesencephalon. Development 135, 3459–3470 (2008).

    CAS  PubMed  Google Scholar 

  252. Andersson, E. et al. Identification of intrinsic determinants of midbrain dopamine neurons. Cell 124, 393–405 (2006).

    CAS  PubMed  Google Scholar 

  253. Nishiyama, A. et al. Uncovering early response of gene regulatory networks in ESCs by systematic induction of transcription factors. Cell Stem Cell 5, 420–433 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  254. Hobert, O., Carrera, I. & Stefanakis, N. The molecular and gene regulatory signature of a neuron. Trends Neurosci. 33, 435–445 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  255. Gaspard, N. & Vanderhaeghen, P. Mechanisms of neural specification from embryonic stem cells. Curr. Opin. Neurobiol. 20, 37–43 (2010).

    CAS  PubMed  Google Scholar 

  256. Gaspard, N. et al. An intrinsic mechanism of corticogenesis from embryonic stem cells. Nature 455, 351–357 (2008).

    CAS  PubMed  Google Scholar 

  257. Zhang, X. et al. Pax6 is a human neuroectoderm cell fate determinant. Cell Stem Cell 7, 90–100 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  258. Kriegstein, A., Noctor, S. & Martinez-Cerdeno, V. Patterns of neural stem and progenitor cell division may underlie evolutionary cortical expansion. Nat. Rev. Neurosci. 7, 883–890 (2006).

    CAS  PubMed  Google Scholar 

  259. Murry, C.E. & Keller, G. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell 132, 661–680 (2008).

    CAS  PubMed  Google Scholar 

  260. Elkabetz, Y. et al. Human ES cell-derived neural rosettes reveal a functionally distinct early neural stem cell stage. Genes Dev. 22, 152–165 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  261. Chambers, S.M. et al. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat. Biotechnol. 27, 275–280 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  262. Koch, P., Opitz, T., Steinbeck, J.A., Ladewig, J. & Brustle, O. A rosette-type, self-renewing human ES cell-derived neural stem cell with potential for in vitro instruction and synaptic integration. Proc. Natl. Acad. Sci. USA 106, 3225–3230 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  263. Hu, B.Y. et al. Neural differentiation of human induced pluripotent stem cells follows developmental principles but with variable potency. Proc. Natl. Acad. Sci. USA 107, 4335–4340 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  264. Lee, G. et al. Modelling pathogenesis and treatment of familial dysautonomia using patient-specific iPSCs. Nature 461, 402–406 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  265. Marchetto, M.C. et al. A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell 143, 527–539 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  266. Brennand, K.J. et al. Modelling schizophrenia using human induced pluripotent stem cells. Nature 473, 221–225 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  267. Nguyen, H.N. et al. LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress. Cell Stem Cell 8, 267–280 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  268. Carvajal-Vergara, X. et al. Patient-specific induced pluripotent stem-cell-derived models of LEOPARD syndrome. Nature 465, 808–812 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  269. Yazawa, M. et al. Using induced pluripotent stem cells to investigate cardiac phenotypes in Timothy syndrome. Nature 471, 230–234 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  270. Angel, M. & Yanik, M.F. Innate immune suppression enables frequent transfection with RNA encoding reprogramming proteins. PLoS ONE 5, e11756 (2010).

    PubMed  PubMed Central  Google Scholar 

  271. Warren, L. et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 7, 618–630 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  272. Laiosa, C.V. et al. Reprogramming of committed T cell progenitors to macrophages and dendritic cells by C/EBP alpha and PU.1 transcription factors. Immunity 25, 731–744 (2006).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank the many colleagues who have contributed to this work through many insightful discussions on the topic over the last year. These include T. Graf, F. Guillemot, M. Götz, J. Crabtree, A. Smith, R. Jaenisch, J. Wysocka, H. Blau, H. Chang, A. Kriegstein and I. Weissman. M.W. is a New York Stem Cell Foundation (NYSCF) Robertson Investigator, and our research is also supported by grants from the US National Institutes of Health, the Department of Defense, the California Institute of Regenerative Medicine, the BioX program at Stanford, the Baxter Foundation and the Stinehart-Reed Foundation. T.V. is supported by the Ruth and Robert Halperin Stanford Graduate Fellowship. We apologize to numerous authors whose valuable contributions we were unable to cite due to space constraints.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Marius Wernig.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Vierbuchen, T., Wernig, M. Direct lineage conversions: unnatural but useful?. Nat Biotechnol 29, 892–907 (2011). https://doi.org/10.1038/nbt.1946

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.1946

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