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:

Derive and conquer: sourcing and differentiating stem cells for therapeutic applications

Key Points

  • Human embryonic stem cells (ESCs) hold great promise for regenerative medicine. However, many technical hurdles and unanswered questions remain before this research can be safely translated into real treatments in the clinic. These range from cell sourcing and the efficient production of differentiated cell derivatives to the ethical issues related to the use of human embryos.

  • Safety comes first when it comes to human clinical trials. This requires human ESC derivatives to be free of human and animal pathogens, particularly if feeder cells and/or other animal products are used for co-culture. The derivatives also have to undergo extensive safety testing to ensure that they are non-tumorigenic.

  • Immune rejection is another major obstacle associated with the transplantation of cells and tissues. Several strategies are currently being explored to generate histocompatible human ESC derivatives, including somatic cell nuclear transfer, parthenogenesis and cellular reprogramming.

  • The destruction of human embryos to generate ESCs remains a major source of ethical controversy. Several strategies have been proposed to solve this problem. These include the use of cell reprogramming, parthenogenesis, altered nuclear transfer and the use of biopsied cells removed from embryos using a technique similar to pre-implantation genetic diagnosis.

  • For clinical studies, large enough quantities of the appropriate replacement cell type will need to be generated under well-defined and reproducible conditions using traceable reagents. Purity, yield and functionality of the cells will also need to be optimized.

  • Non-embryonic tissues may also be an important source of stem cells. Multipotent and pluripotent cells have been successfully isolated from bone marrow, adipose tissue, skin, teeth, testes, amniotic fluid and umbilical-cord blood, among others. These stem cells have been shown to differentiate into a variety of important cell types, and have the advantage of being histocompatible with the individual from which they were derived.

Abstract

Although great progress has been made in the isolation and culture of stem cells, the future of stem-cell-based therapies and their productive use in drug discovery and regenerative medicine depends on two key factors: finding reliable sources of multipotent and pluripotent cells and the ability to control their differentiation to generate desired derivatives. It is essential for clinical applications to establish reliable sources of pathogen-free human embryonic stem cells (ESCs) and develop suitable differentiation techniques. Here, we address some of the problems associated with the sourcing of human ESCs and discuss the current status of stem-cell differentiation technology.

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: Possible ways to generate immune-compatible derivatives of pluripotent cells.
Figure 2: In vitro avenues to differentiation of embryonic stem cells (ESCs).

Similar content being viewed by others

References

  1. Martin, G. R. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl Acad. Sci. USA 78, 7634–7638 (1981).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Evans, M. J. & Kaufman, M. H. Establishment in culture of pluripotential cells from mouse embryos. Nature 292, 154–156 (1981).

    CAS  PubMed  Google Scholar 

  3. Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 1145–1147 (1998).

    CAS  PubMed  Google Scholar 

  4. Shamblott, M. J. et al. Derivation of pluripotent stem cells from cultured human primordial germ cells. Proc. Natl Acad. Sci. USA 95, 13726–13731 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Ben-Hur, T. et al. Transplantation of human embryonic stem cell-derived neural progenitors improves behavioral deficit in Parkinsonian rats. Stem Cells 22, 1246–1255 (2004).

    PubMed  Google Scholar 

  6. Roy, N. S. et al. Functional engraftment of human ES cell-derived dopaminergic neurons enriched by coculture with telomerase-immortalized midbrain astrocytes. Nature Med. 12, 1259–1268 (2006).

    CAS  PubMed  Google Scholar 

  7. Kehat, I. et al. Electromechanical integration of cardiomyocytes derived from human embryonic stem cells. Nature Biotech. 22, 1282–1289 (2004).

    CAS  Google Scholar 

  8. Laflamme, M. A. et al. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nature Biotech. 25, 1015–1024 (2007). A demonstration of the functionality of ESC derivatives from the mesoderm lineage.

    CAS  Google Scholar 

  9. Xie, C. Q. et al. Transplantation of human undifferentiated embryonic stem cells into a myocardial infarction rat model. Stem Cells Dev. 16, 25–29 (2007).

    CAS  PubMed  Google Scholar 

  10. Haruta, M. et al. In vitro and in vivo characterization of pigment epithelial cells differentiated from primate embryonic stem cells. Invest. Ophthalmol. Vis. Sci. 45, 1020–1025 (2004).

    PubMed  Google Scholar 

  11. Lund R. D. et al. Human embryonic stem cell-derived cells rescue visual function in dystrophic RCS rats. Cloning Stem Cells 8, 189–199 (2006). A demonstration of the functionality of ESC derivatives from the ectoderm lineage.

    CAS  PubMed  Google Scholar 

  12. Zambrowicz, B. P. & Sands, A. T. Knockouts model the 100 best-selling drugs — will they model the next 100? Nature Rev. Drug Discov. 2, 38–51 (2003).

    CAS  Google Scholar 

  13. Rosenthal, N. & Brown, S. The mouse ascending: perspectives for human-disease models. Nature Cell Biol. 9, 993–999 (2007).

    CAS  PubMed  Google Scholar 

  14. McNeish J. Embryonic stem cells in drug discovery. Nature Rev. Drug Discov. 3, 70–80 (2004).

    CAS  Google Scholar 

  15. Verlinsky, Y. et al. Human embryonic stem cell lines with genetic disorders. Reprod. Biomed. Online 10, 105–110 (2005).

    CAS  PubMed  Google Scholar 

  16. Weber, D. J. Manufacturing considerations for clinical uses of therapies derived from stem cells. Methods Enzymol. 420, 410–430 (2006).

    CAS  PubMed  Google Scholar 

  17. Stojkovic, M. An autogeneic feeder cell system that efficiently supports growth of undifferentiated human embryonic stem cells. Stem Cells 23, 306–314 (2005).

    CAS  PubMed  Google Scholar 

  18. Genbacev, O. et al. Serum-free derivation of human embryonic stem cell lines on human placental fibroblast feeders. Fertil. Steril. 83, 1517–1529 (2005).

    PubMed  Google Scholar 

  19. Simon, C. et al. First derivation in Spain of human embryonic stem cell lines: use of long-term cryopreserved embryos and animal-free conditions. Fertil. Steril. 83, 246–249 (2005).

    PubMed  Google Scholar 

  20. Klimanskaya, I. et al. Human embryonic stem cells derived without feeder cells. Lancet 365, 1636–1641 (2005).

    CAS  PubMed  Google Scholar 

  21. Ludwig, T. E. et al. Derivation of human embryonic stem cells in defined conditions. Nature Biotech. 24, 185–187 (2006).

    CAS  Google Scholar 

  22. Amit, M. & Itskovitz-Eldor J. Feeder-free culture of human embryonic stem cells. Methods Enzymol. 420, 37–49 (2006).

    CAS  PubMed  Google Scholar 

  23. Ludwig, T. E. et al. Feeder-independent culture of human embryonic stem cells. Nature Methods 3, 637–646 (2006).

    CAS  PubMed  Google Scholar 

  24. Pyle, A. D., Lock, L. F. & Donovan, P. J. Neurotrophins mediate human embryonic stem cell survival. Nature Biotech. 24, 344–350 (2006).

    CAS  Google Scholar 

  25. Lu, J., Hou, R., Booth, C. J., Yang, S. H. & Snyder, M. Defined culture conditions of human embryonic stem cells. Proc. Natl Acad. Sci. USA 103, 5688–5693 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Arnhold, S., Klein, H., Semkova, I., Addicks, K. & Schraermeyer, U. Neurally selected embryonic stem cells induce tumor formation after long-term survival following engraftment into the subretinal space. Invest. Ophthalmol. Vis. Sci. 45, 4251–4255 (2004). This paper shows that even differentiated ESCs can cause tumours.

    PubMed  Google Scholar 

  27. Wernig, M. et al. Functional integration of embryonic stem cell-derived neurons in vivo. J. Neurosci. 24, 5258–5268 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Hwang, Y. Y. et al. Patient-specific embryonic stem cells derived from human SCNT blastocysts. Science 308, 1777–1783 (2005).

    CAS  PubMed  Google Scholar 

  29. Hwang, W. S. et al. Evidence of a pluripotent human embryonic stem cell line derived from a cloned blastocyst. Science 303, 1669–1674 (2004).

    CAS  PubMed  Google Scholar 

  30. Brambrink, T., Hochedlinger, K., Bell, G. & Jaenisch, R. ES cells derived from cloned and fertilized blastocysts are transcriptionally and functionally indistinguishable. Proc. Natl Acad. Sci. USA 103, 933–938 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Hochedlinger, K. & Jaenisch, R. Nuclear transplantation, embryonic stem cells, and the potential for cell therapy. N. Engl. J. Med. 349, 275–286 (2003).

    CAS  PubMed  Google Scholar 

  32. Kohda, T. et al. Variation in gene expression and aberrantly regulated chromosome regions in cloned mice. Biol. Reprod. 73, 1302–1311 (2005).

    CAS  PubMed  Google Scholar 

  33. Humpherys, D. et al. Abnormal gene expression in cloned mice derived from embryonic stem cell and cumulus cell nuclei. Proc. Natl Acad. Sci. USA 99, 12889–12894 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Bortvin, A. et al. Incomplete reactivation of Oct4-related genes in mouse embryos cloned from somatic nuclei. Development 130, 1673–1680 (2003).

    CAS  PubMed  Google Scholar 

  35. Boiani, M., Eckardt, S., Scholer, H. R. & McLaughlin, K. J. Oct4 distribution and level in mouse clones: consequences for pluripotency. Genes Dev. 16, 1209–1219 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Smith, S. L. et al. Global gene expression profiles reveal significant nuclear reprogramming by the blastocyst stage after cloning. Proc. Natl Acad. Sci. USA 102, 17582–17587 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Rhind, S. M. et al. Cloned lambs — lessons from pathology. Nature Biotech. 21, 744–745 (2003).

    CAS  Google Scholar 

  38. Cibelli, J. B. et al. Parthenogenetic stem cells in nonhuman primates. Science 295, 819 (2002).

    CAS  PubMed  Google Scholar 

  39. Lin, H. et al. Multilineage potential of homozygous stem cells derived from metaphase II oocytes. Stem Cells 2, 152–161 (2003).

    Google Scholar 

  40. Revazova, E. S. et al. Patient-specific stem cell lines derived from human parthenogenetic blastocysts. Cloning Stem Cells 9, 432–449 (2007).

    CAS  PubMed  Google Scholar 

  41. Kitai, K. et al. Recombination signatures distinguish embryonic stem cells derived by parthenogenesis and somatic cell nuclear transfer. Cell Stem Cell 1, 346–352 (2007).

    Google Scholar 

  42. Hakelien, A. M., Landsverk, H. B., Robl, J. M., Skalhegg, B. S. & Collas, P. Reprogramming fibroblasts to express T-cell functions using cell extracts. Nature Biotech. 20, 460–466 (2002).

    CAS  Google Scholar 

  43. Gaustad, K. G., Boquest, A. C., Anderson, B. E., Gerdes, A. M. & Collas, P. Differentiation of human adipose tissue stem cells using extracts of rat cardiomyocytes. Biochem. Biophys. Res. 314, 420–427 (2004).

    CAS  Google Scholar 

  44. Qin, M., Tai, G., Collas, P., Polak, J. M. & Bishop, A. E. Cell extract-derived differentiation of embryonic stem cells. Stem Cells 23, 712–718 (2005).

    CAS  PubMed  Google Scholar 

  45. Taranger, C. K. et al. Induction of dedifferentiation, genomewide transcriptional programming, and epigenetic reprogramming by extracts of carcinoma and embryonic stem cells. Mol. Biol. Cell 16, 5719–5735. (2005).

    Google Scholar 

  46. 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 

  47. 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 

  48. Strelchenko, N. et al. Reprogramming of human somatic cells by embryonic stem cell cytoplast. Reprod. Biomed. Online 12, 107–111 (2006).

    PubMed  Google Scholar 

  49. Do, J. T. & Scholer, H. R. Nuclei of embryonic stem cells reprogram somatic cells. Stem Cells 22, 941–949 (2004).

    CAS  PubMed  Google Scholar 

  50. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  52. Wernig, M. et al. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448, 318–324 (2007). Together with references 40, 48 and 49, this paper describes transdifferentiation and reprogramming approaches and advances.

    CAS  PubMed  Google Scholar 

  53. 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 

  54. Sipione, S., Eshpeter, A., Lyon, J. G., Korbutt, G. S. & Bleackley, R. C. Insulin expressing cells from differentiated embryonic stem cells are not β cells. Diabetologia 47, 499–508 (2004).

    CAS  PubMed  Google Scholar 

  55. Kania, G., Blyszczuk, P. & Wobus, A. M. The generation of insulin-producing cells from embryonic stem cells — a discussion of controversial findings. Int. J. Dev. Biol. 48, 1061–1064. (2004). A comparison of various strategies used to generate pancreatic cells with respect to protocols and differentiation factors, which aims to give an explanation of the contradictory findings that have been published.

    PubMed  Google Scholar 

  56. Wei, H., Juhasz, O., Li, J., Tarasova, Y. S. & Boheler, K. R. Embryonic stem cells and cardiomyocyte differentiation: phenotypic and molecular analyses. J. Cell. Mol. Med. 9, 804–817 (2005).

    PubMed  PubMed Central  Google Scholar 

  57. Teramoto, K. et al. Hepatocyte differentiation from embryonic stem cells and umbilical cord blood cells. J. Hepatobiliary Pancreat. Surg. 12, 196–202 (2005).

    PubMed  Google Scholar 

  58. Olsen, A. L., Stachura, D. L. & Weiss, M. J. Designer blood: creating hematopoietic lineages from embryonic stem cells. Blood 107, 1265–1275 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Johansson, B. M. & Wiles, M. V. Evidence for involvement of activin A and bone morphogenetic protein 4 in mammalian mesoderm and hematopoietic development. Mol. Cell Biol. 15, 141–151 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Suzuki, A. et al. C. Nanog binds to Smad1 and blocks bone morphogenetic protein-induced differentiation of embryonic stem cells. Proc. Natl Acad. Sci. USA 103, 10294–10299 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Wiles, M. V. & Johansson, B. M. Analysis of factors controlling primary germ layer formation and early hematopoiesis using embryonic stem cell in vitro differentiation. Leukemia 11 (Suppl. 3), 454–456 (1997).

    PubMed  Google Scholar 

  62. Finley, M. F., Devata, S. & Huettner, J. E. BMP-4 inhibits neural differentiation of murine embryonic stem cells. J. Neurobiol. 40, 271–287 (1999).

    CAS  PubMed  Google Scholar 

  63. Kawasaki, H. et al. Induction of midbrain dopaminergic neurons from ES cells by stromal cell-derived inducing activity. Neuron 28, 31–40 (2000).

    CAS  PubMed  Google Scholar 

  64. Rodda, S. J., Kavanagh, S. J., Rathjen, J. & Rathjen, P. D. Embryonic stem cell differentiation and the analysis of mammalian development. Int. J. Dev. Biol. 46, 449–458 (2002).

    CAS  PubMed  Google Scholar 

  65. Sonntag, K. C. et al. Enhanced yield of neuroepithelial precursors and midbrain-like dopaminergic neurons from human embryonic stem cells using the bone morphogenic protein antagonist noggin. Stem Cells 25, 411–418 (2007).

    CAS  PubMed  Google Scholar 

  66. Pera, M. F. et al. Regulation of human embryonic stem cell differentiation by BMP-2 and its antagonist noggin. J. Cell. Sci. 117, 1269–1280 (2004).

    CAS  PubMed  Google Scholar 

  67. Gratsch, T. E. & O'Shea, K. S. Noggin and chordin have distinct activities in promoting lineage commitment of mouse embryonic stem (ES) cells. Dev. Biol. 245, 83–94 (2002).

    CAS  PubMed  Google Scholar 

  68. Tropepe, V. et al. Direct neural fate specification from embryonic stem cells: a primitive mammalian neural stem cell stage acquired through a default mechanism. Neuron 30, 65–78 (2001).

    CAS  PubMed  Google Scholar 

  69. Lei, T. et al. Xeno-free derivation and culture of human embryonic stem cells: current status, problems and challenges. Cell Res. 17, 682–688 (2007).

    CAS  PubMed  Google Scholar 

  70. Okabe, S., Forsberg-Nilsson, K., Spiro, A. C., Segal, M. & McKay, R. D. Development of neuronal precursor cells and functional postmitotic neurons from embryonic stem cells in vitro. Mech. Dev. 59, 89–102 (1996).

    CAS  PubMed  Google Scholar 

  71. Lee, S. H., Lumelsky, N., Studer, L., Auerbach, J. M. & McKay, R. D. Efficient generation of midbrain and hindbrain neurons from mouse embryonic stem cells. Nature Biotech. 18, 675–679 (2000).

    CAS  Google Scholar 

  72. Lumelsky, N. et al. Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic islets Science 292, 1389–1394 (2001).

    CAS  PubMed  Google Scholar 

  73. Rajagopal, J., Anderson, W. J., Kume, S., Martinez, O. I. & Melton, D. A. Insulin staining of ES cell progeny from insulin uptake. Science 299, 363 (2003).

    PubMed  Google Scholar 

  74. Hori, Y. et al. Growth inhibitors promote differentiation of insulin-producing tissue from embryonic stem cells. Proc. Natl Acad. Sci. USA 99, 16105–16110 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Hansson, M. et al. Artifactual insulin release from differentiated embryonic stem cells. Diabetes 53, 2603–2609 (2004).

    CAS  PubMed  Google Scholar 

  76. D'Amour, K. A. et al. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nature Biotech. 1481–1483 (2006). A demonstration of the functionality of ESC derivatives from the endoderm lineage.

  77. Shim, J. H. et al. Directed differentiation of human embryonic stem cells towards a pancreatic cell fate. Diabetologia 50, 1228–1238 (2007).

    CAS  PubMed  Google Scholar 

  78. Phillips, B. W. et al. Directed differentiation of human embryonic stem cells into the pancreatic endocrine lineage. Stem Cells Dev. 16, 561–578 (2007).

    CAS  PubMed  Google Scholar 

  79. Doetschman, T. C., Eistetter, H., Katz, M., Schmidt, W. & Kemler, R. The in vitro development of blastocyst-derived embryonic stem cell lines: formation of visceral yolk sac, blood islands and myocardium. J. Embryol. Exp. Morphol. 87, 27–45 (1985).

    CAS  PubMed  Google Scholar 

  80. Boheler, K. R. et al. Differentiation of pluripotent embryonic stem cells into cardiomyocytes. Circ. Res. 91, 189–201 (2002).

    CAS  PubMed  Google Scholar 

  81. Kehat, I. et al. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J. Clin. Invest. 108, 407–414 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Sachinidis, A. et al. Identification of plateled-derived growth factor-BB as cardiogenesis-inducing factor in mouse embryonic stem cells under serum-free conditions. Cell. Physiol. Biochem. 13, 423–429 (2003).

    CAS  PubMed  Google Scholar 

  83. Srivastava, D. & Ivey, K. N. Potential of stem-cell-based therapies for heart disease. Nature 441, 1097–1099 (2006).

    CAS  PubMed  Google Scholar 

  84. Sachinidis, A. et al. Identification of small signalling molecules promoting cardiac-specific differentiation of mouse embryonic stem cells. Cell. Physiol. Biochem. 18, 303–314 (2006).

    CAS  PubMed  Google Scholar 

  85. Buggisch, M. et al. Stimulation of ES-cell-derived cardiomyogenesis and neonatal cardiac cell proliferation by reactive oxygen species and NADPH oxidase. J. Cell Sci. 120, 885–894 (2007).

    CAS  PubMed  Google Scholar 

  86. Zhao, Y. et al. Dysregulation of cardiogenesis, cardiac conduction, and cell cycle in mice lacking miRNA-1-2. Cell 29, 303–317 (2007).

    Google Scholar 

  87. van Rooij, E. et al. Control of stress-dependent cardiac growth and gene expression by a microRNA. Science 316, 575–579 (2007).

    CAS  PubMed  Google Scholar 

  88. Lough, J. & Sugi, Y. Endoderm and heart development. Dev. Dyn. 217, 327–342 (2000).

    CAS  PubMed  Google Scholar 

  89. Mummery, C. et al. Differentiation of human embryonic stem cells to cardiomyocytes: role of coculture with visceral endoderm-like cells. Circulation 107, 2733–2740 (2003).

    CAS  PubMed  Google Scholar 

  90. Passier, R. et al. Increased cardiomyocyte differentiation from human embryonic stem cells in serum-free cultures. Stem Cells 23, 772–780 (2005).

    CAS  PubMed  Google Scholar 

  91. Kumar, D., Kamp, T. J. & LeWinter, M. M. Embryonic stem cells: differentiation into cardiomyocytes and potential for heart repair and regeneration. Coron. Artery Dis. 16, 111–116 (2005).

    PubMed  Google Scholar 

  92. Caspi, O. & Gepstein, L. Potential applications of human embryonic stem cell-derived cardiomyocytes. Ann. NY Acad. Sci. 1015, 285–298 (2004).

    PubMed  Google Scholar 

  93. Dolnikov, K. et al. Functional properties of human embryonic stem cell-derived cardiomyocytes. Ann. NY Acad. Sci. 1047, 66–75 (2005).

    CAS  PubMed  Google Scholar 

  94. Passier, R. & Mummery, C. Cardiomyocyte differentiation from embryonic and adult stem cells. Curr. Opin. Biotechnol. 1, 498–502 (2005).

    Google Scholar 

  95. Menard, C. et al. Transplantation of cardiac-committed mouse embryonic stem cells to infarcted sheep myocardium: a preclinical study. Lancet 366, 1005–1012 (2005).

    PubMed  Google Scholar 

  96. Xue, T. et al. Functional integration of electrically active cardiac derivatives from genetically engineered human embryonic 28 stem cells with quiescent recipient ventricular cardiomyocytes: insights into the development of cell-based pacemakers. Circulation 111, 11–20 (2005).

    PubMed  Google Scholar 

  97. Kofidis, T. et al. Allopurinol/uricase and ibuprofen enhance engraftment of cardiomyocyte-enriched human embryonic stem cells and improve cardiac function following myocardial injury. Eur. J. Cardiothorac. Surg. 29, 50–55 (2006).

    PubMed  Google Scholar 

  98. Smukler, S. R., Runciman, S. B., Xu, S. & van der Kooy, D. Embryonic stem cells assume a primitive neural stem cell fate in the absence of extrinsic influences. J. Cell Biol. 172, 79–90 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Ying, Q. L., Stavridis, M., Griffiths, D., Li, M. & Smith, A. Conversion of embryonic stem cells into neuroectodermal precursors in adherent monoculture. Nature Biotech. 21, 183–186 (2003).

    CAS  Google Scholar 

  100. Munoz-Sanjuan, I. & Brivanlou, A. H. Neural induction, the default model and embryonic stem cells. Nature Rev. Neurosci. 3, 271–280 (2002).

    CAS  Google Scholar 

  101. Carpenter, M. K. et al. Enrichment of neurons and neural precursors from human embryonic stem cells. Exp. Neurol. 172, 383–397 (2001).

    CAS  PubMed  Google Scholar 

  102. Reubinoff, B. E. et al. Neural progenitors from human embryonic stem cells. Nature Biotech. 19, 1134–1140 (2001).

    CAS  Google Scholar 

  103. Meyer, J. S., Katz, M. L., Maruniak, J. A. & Kirk, M. D. Embryonic stem cell-derived neural progenitors incorporate into degenerating retina and enhance survival of host photoreceptors. Stem Cells 24, 274–283 (2006).

    PubMed  Google Scholar 

  104. Takagi, Y. et al. Dopaminergic neurons generated from monkey embryonic stem cells function in a Parkinson primate model. J. Clin. Invest. 115, 102–109 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Morizane, A. et al. Generation of graftable dopaminergic neuron progenitors from mouse ES cells by a combination of coculture and neurosphere methods. J. Neurosci. Res. 83, 1015–1027 (2006).

    CAS  PubMed  Google Scholar 

  106. Ben-Hur, T. Human embryonic stem cells for neuronal repair. Isr. Med. Assoc. J. 8, 122–126 (2006).

    PubMed  Google Scholar 

  107. Kim, J. H. et al. Dopamine neurons derived from embryonic stem cells function in an animal model of Parkinson's disease. Nature 418, 50–56 (2002).

    CAS  PubMed  Google Scholar 

  108. Kawasaki, H. et al. Generation of dopaminergic neurons and pigmented epithelia from primate ES cells by stromal cell-derived inducing activity. Proc. Natl Acad. Sci. USA 99, 1580–1585 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Rolletschek, A. et al. Differentiation of embryonic stem cell-derived dopaminergic neurons is enhanced by survival-promoting factors. Mech. Dev. 105, 93–104 (2001).

    CAS  PubMed  Google Scholar 

  110. Perrier, A. L. et al. Derivation of midbrain dopamine neurons from human embryonic stem cells. Proc. Natl Acad. Sci. USA 101, 12543–12548 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Zeng, X. et al. Dopaminergic differentiation of human embryonic stem cells. Stem Cells 22, 925–940 (2004).

    CAS  PubMed  Google Scholar 

  112. Bjorklund, L. M. et al. Embryonic stem cells develop into functional dopaminergic neurons after transplantation in a Parkinson rat model. 99, 2344–2349 (2002).

  113. Nishimura, F. et al. Potential use of embryonic stem cells for the treatment of mouse parkinsonian models: improved behavior by transplantation of in vitro differentiated dopaminergic neurons from embryonic stem cells. Stem Cells 21, 171–180 (2003).

    PubMed  Google Scholar 

  114. Yoshizaki, T. et al. Isolation and transplantation of dopaminergic neurons generated from mouse embryonic stem cells. Neurosci. Lett. 363, 33–37 (2004).

    CAS  PubMed  Google Scholar 

  115. Barberi, T. et al. Neural subtype specification of fertilization and nuclear transfer embryonic stem cells and application in parkinsonian mice. Nature Biotech. 21, 1200–1207 (2003).

    CAS  Google Scholar 

  116. Inden, M. et al. Transplantation of mouse embryonic stem cell-derived neurons into the striatum, subthalamic nucleus and substantia nigra, and behavioral recovery inhemiparkinsonian rats. Neurosci. Lett. 387, 151–156 (2005).

    CAS  PubMed  Google Scholar 

  117. Chiba, S., Iwasaki, Y., Sekino, H. & Suzuki, N. Transplantation of motoneuron-enriched neural cells derived from mouse embryonic stem cells improves motor function of hemiplegic mice. Cell Transplant. 12, 457–468 (2003).

    PubMed  Google Scholar 

  118. Ikeda, R. et al. Transplantation of motoneurons derived from MASH1-transfected mouse ES cells reconstitutes neural networks and improves motor function in hemiplegic mice. Exp. Neurol. 189, 280–292 (2004).

    CAS  PubMed  Google Scholar 

  119. Faulkner, J. & Keirstead, H. S. Human embryonic stem cell-derived oligodendrocyte progenitors for the treatment of spinal cord injury. Transpl. Immunol. 15, 131–142 (2005).

    CAS  PubMed  Google Scholar 

  120. McDonald, J. W. & Howard, M. J. Repairing the damaged spinal cord: a summary of our early success with embryonic stem cell transplantation and remyelination. Prog. Brain Res. 137, 299–309 (2002).

    PubMed  Google Scholar 

  121. McDonald, J. W. et al. Repair of the injured spinal cord and the potential of embryonic stem cell transplantation. 21, 383–393 (2004).

  122. Myckatyn, T. M., Mackinnon, S. E. & McDonald, J. W. Stem cell transplantation and other novel techniques for promoting recovery from spinal cord injury. Transpl. Immunol. 12, 343–358 (2004).

    CAS  PubMed  Google Scholar 

  123. Takagi, Y. et al. Survival and differentiation of neural progenitor cells derived from embryonic stem cells and transplanted into ischemic brain. J. Neurosurg. 103, 304–310 (2005).

    PubMed  Google Scholar 

  124. Hayashi, J. et al. Primate embryonic stem cell-derived neuronal progenitors transplanted into ischemic brain. J. Cereb. Blood Flow Metab. 26, 906–914 (2006).

    PubMed  Google Scholar 

  125. Hoane, M. R. et al. Transplantation of neuronal and glial precursors dramatically improves sensorimotor function but not cognitive function in the traumatically injured brain. J. Neurotrauma 21, 163–174 (2004).

    PubMed  Google Scholar 

  126. Fuhrmann, S., Levine, E. M. & Reh, T. A. Extraocular mesenchyme patterns the optic vesicle during early eye development in the embryonic chick. Development 127, 4599–4609 (2000).

    CAS  PubMed  Google Scholar 

  127. Chow, R. L. & Lang, R. A. Early eye development in vertebrates. Annu. Rev. Cell Dev. Biol. 17, 255–296. (2001).

    CAS  PubMed  Google Scholar 

  128. Morizane, A., Takahashi, J., Takagi, Y., Sasai, Y. & Hashimoto, N. Optimal conditions for in vivo induction of dopaminergic neurons from embryonic stem cells through stromal cell-derived inducing activity. J. Neurosci. Res. 69, 934–939 (2002).

    CAS  PubMed  Google Scholar 

  129. Klimanskaya, I. et al. Derivation and comparative assessment of retinal pigment epithelium from human embryonic stem cells using transcriptomics. Cloning Stem Cells 6, 217–245 (2004).

    CAS  PubMed  Google Scholar 

  130. Chung, Y. et al. Embryonic and extraembryonic stem cell lines derived from single mouse blastomeres. Nature 439, 216–219 (2006).

    CAS  PubMed  Google Scholar 

  131. Klimanskaya, I., Chung, Y., Becker, S., Lu, S. J. & Lanza, R. Human embryonic stem cell lines derived from single blastomeres. Nature 444, 481–485 (2006). This is a possible alternative approach to generate human ESCs without embryo destruction.

    CAS  PubMed  Google Scholar 

  132. Meissner, A. & Jaenisch, R. Generation of nuclear transfer-derived pluripotent ES cells from cloned Cdx2-deficient blastocysts. Nature 439, 212–215 (2006).

    CAS  PubMed  Google Scholar 

  133. Zhang, X. et al. Derivation of human embryonic stem cells from developing and arrested embryos. Stem Cells 24, 2669–2676 (2006).

    CAS  PubMed  Google Scholar 

  134. Schwartz, P. H. & Rae, S. B. An approach to the ethical donation of human embryos for harvest of stem cells. Reprod. Biomed. Online 12, 771–775 (2006).

    PubMed  Google Scholar 

  135. Quaini, F. et al. Chimerism of the transplanted heart. N. Engl. J. Med. 346, 5–15 (2002).

    PubMed  Google Scholar 

  136. Muller, P. et al. Cardiomyocytes of noncardiac origin in myocardial biopsies of human transplanted hearts. Circulation 106, 31–35 (2002).

    PubMed  Google Scholar 

  137. Urbanek, K. et al. Cardiac stem cells possess growth factor-receptor systems that after activation regenerate the infracted myocardium, improving ventricular function and long-term survival. Circ. Res. 97, 663–673 (2005).

    CAS  PubMed  Google Scholar 

  138. Freitas, C. S. & Dalmau, S. R. Multiple sources of non-embryonic multipotent stem cells: processed lipoaspirates and dermis as promising alternatives to bone-marrow-derived cell therapies. Cell Tissue Res. 325, 403–411 (2006).

    PubMed  Google Scholar 

  139. Gronthos, S. et al. Molecular and cellular characterisation of highly purified stromal stem cells derived from human bone marrow. J. Cell Sci. 116, 1827–1835 (2003).

    CAS  PubMed  Google Scholar 

  140. D'Ippolito, G. et al. Marrow-isolated adult multilineage inducible (MIAMI) cells, a unique population of postnatal young and old human cells with extensive expansion and differentiation potential. J. Cell Sci. 117, 2971–2981 (2004).

    CAS  PubMed  Google Scholar 

  141. Pomerantz, J. & Blau, H. M. Nuclear reprogramming: a key to stem cell function in regenerative medicine. Nature Cell Biol. 6, 810–816 (2004).

    CAS  PubMed  Google Scholar 

  142. Guan, K. et al. Pluripotency of spermatogonial stem cells from adult mouse testis. Nature 440, 1199–1203 (2006). Gives an example of an alternative source of pluripotent cells.

    CAS  PubMed  Google Scholar 

  143. Jung, J. G. et al. Identification, culture, and characterization of germline stem cell-like cells in chicken testes. Biol. Reprod. 76, 173–182 (2007).

    CAS  PubMed  Google Scholar 

  144. Waters, J. M., Richardson, G. D. & Jahoda, C. A. Hair follicle stem cells. Semin. Cell Dev. Biol. 18, 245–254 (2007).

    CAS  PubMed  Google Scholar 

  145. Fuchs, E. Scratching the surface of skin development. Nature 445, 834–842 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Cotsarelis, G. Epithelial stem cells: a folliculocentric view. J. Invest. Dermatol. 126, 1459–1468 (2006).

    CAS  PubMed  Google Scholar 

  147. Moore, K. A. & Lemischka, I. R. Stem cells and their niches. Science 311, 1880–1885 (2006).

    CAS  PubMed  Google Scholar 

  148. Roche, R., Hoareau, L., Mounet, F. & Festy, F. Adult stem cells for cardiovascular diseases: the adipose tissue potential. Expert Opin. Biol. Ther. 7, 791–798 (2007).

    CAS  PubMed  Google Scholar 

  149. Gimble, J. M., Katz, A. J. & Bunnell, B. A. Adipose-derived stem cells for regenerative medicine. Circ. Res. 100, 1249–1260 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Zhang, W., Walboomers, X. F., Shi, S., Fan, M. & Jansen, J. A. Multilineage differentiation potential of stem cells derived from human dental pulp after cryopreservation. Tissue Eng. 12, 2813–2823 (2006).

    CAS  PubMed  Google Scholar 

  151. Ballini, A. et al. In vitro stem cell cultures from human dental pulp and periodontal ligament: new prospects in dentistry. Int. J. Immunopathol. Pharmacol. 20, 9–16 (2007).

    CAS  PubMed  Google Scholar 

  152. Miura, M. et al. SHED: stem cells from human exfoliated deciduous teeth. Proc. Natl Acad. Sci. USA 100, 5807–5812 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. McGuckin, C. P. et al. Production of stem cells with embryonic characteristics from human umbilical cord blood. Cell Prolif. 38, 245–255 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. De Coppi, P. et al. Amniotic fluid and bone marrow derived mesenchymal stem cells can be converted to smooth muscle cells in the cryo-injured rat bladder and prevent compensatory hypertrophy of surviving smooth muscle cells. J. Urol. 177, 369–376 (2007).

    PubMed  Google Scholar 

  155. Ilancheran, S. et al. Stem cells derived from human fetal membranes display multi-lineage differentiation potential. Biol. Reprod. 77, 577–588 (2007).

    CAS  PubMed  Google Scholar 

  156. Mizuseki, K. et al. Generation of neural crest-derived peripheral neurons and floor plate cells from mouse and primate embryonic stem cells. Proc. Natl Acad. Sci. USA 100, 5828–5833 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Lim, U. M., Sidhu, K. S. & Tuch, B. E. Derivation of motor neurons from three clonal human embryonic stem cell lines. Curr. Neurovasc. Res. 3, 281–288 (2006).

    PubMed  Google Scholar 

  158. Soundararajan, P., Miles, G. B., Rubin, L. L., Brownstone, R. M. & Rafuse, V. F. Motoneurons derived from embryonic stem cells express transcription factors and develop phenotypes characteristic of medial motor column neurons. J. Neurosci. 26, 3256–3268 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Willerth, S. M., Faxel, T. E., Gottlieb, D. I. & Sakiyama-Elbert, S. E. The effects of soluble growth factors on embryonic stem cell differentiation inside of fibrin scaffolds. Stem Cells 25, 2235–2244 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Izrael, M. et al. Human oligodendrocytes derived from embryonic stem cells: effect of noggin on phenotypic differentiation in vitro and on myelination in vivo. Mol. Cell Neurosci. 34, 310–323 (2007).

    CAS  PubMed  Google Scholar 

  161. Takenaga, M., Fukumoto, M. & Hori, Y. Regulated nodal signaling promotes differentiation of the definitive endoderm and mesoderm from ES cells. J. Cell Sci. 120, 2078–2090 (2007).

    CAS  PubMed  Google Scholar 

  162. Lu, S. J. et al. Generation of functional hemangioblasts from human embryonic stem cells. Nature Methods 4, 501–509 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Krtolica, A. et al. Disruption of apical-basal polarity of human embryonic stem cells enhances hematoendothelial differentiation. Stem Cells 25, 2215–2223 (2007).

    CAS  PubMed  Google Scholar 

  164. Jiang, J. et al. Generation of insulin-producing islet-like clusters from human embryonic stem cells. Stem Cells 25, 1940–1953 (2007).

    CAS  PubMed  Google Scholar 

  165. Cai, J. et al. Directed differentiation of human embryonic stem cells into functional hepatic cells. Hepatology 45, 1229–1239 (2007).

    CAS  PubMed  Google Scholar 

  166. Soto-Gutierrez, A. et al. Differentiation of mouse embryonic stem cells to hepatocyte-like cells by co-culture with human liver nonparenchymal cell lines. Nature Protoc. 2, 347–356 (2007).

    CAS  Google Scholar 

  167. Chen, Y. et al. Instant hepatic differentiation of human embryonic stem cells using activin A and a deleted variant of HGF. Cell Transplant. 15, 865–871 (2006).

    PubMed  Google Scholar 

  168. Loebel, D. A., Watson, C. M., De Young, R. A. & Tam, P. P. Lineage choice and differentiation in mouse embryos and embryonic stem cells. Dev. Biol. 264, 1–14 (2003).

    CAS  PubMed  Google Scholar 

  169. Nagy, A. & Vintersten, K. Murine embryonic stem cells. Methods Enzymol. 418, 3–21 (2006).

    CAS  PubMed  Google Scholar 

  170. Bryja, V. et al. An efficient method for the derivation of mouse embryonic stem cells. Stem Cells 24, 844–849 (2006).

    PubMed  Google Scholar 

  171. Longo, L., Bygrave, A., Grosveld, F. G. & Pandolfi, P. P. The chromosome make-up of mouse embryonic stem cells is predictive of somatic and germ cell chimaerism. Transgenic Res. 6, 321–328 (1997).

    CAS  PubMed  Google Scholar 

  172. Liu, X. et al. Trisomy eight in ES cells is a common potential problem in gene targeting and interferes with germ line transmission. Dev. Dyn. 209, 85–91 (1997).

    CAS  PubMed  Google Scholar 

  173. Suzuki, H. et al. Germ-line contribution of embryonic stem cells in chimeric mice: influence of karyotype and in vitro differentiation ability. Exp. Anim. 46, 17–23 (1997).

    CAS  PubMed  Google Scholar 

  174. Kondoh, G. et al. Easy assessment of ES cell clone potency for chimeric development and germ-line competency by an optimized aggregation method. J. Biochem. Biophys. Methods 39, 137–142 (1999).

    CAS  PubMed  Google Scholar 

  175. Nichols, J., Chambers, I. & Smith, A. Derivation of germline competent embryonic stem cells with a combination of interleukin-6 and soluble interleukin-6 receptor. Exp. Cell Res. 215, 237–239 (1994).

    CAS  PubMed  Google Scholar 

  176. Rossant, J. Stem cells and lineage development in the mammalian blastocyst. Reprod. Fertil. Dev. 19, 111–118 (2007).

    CAS  PubMed  Google Scholar 

  177. Lakshmipathy, U. et al. Efficient transfection of embryonic and adult stem cells. Stem Cells 22, 531–543 (2004).

    PubMed  Google Scholar 

  178. Smith, A. G. Embryo-derived stem cells: of mice and men. Annu. Rev. Cell Dev. Biol. 17, 435–462 (2001).

    CAS  PubMed  Google Scholar 

  179. Ginis, I. et al. Differences between human and mouse embryonic stem cells. Dev. Biol. 269, 360–380 (2004).

    CAS  PubMed  Google Scholar 

  180. Hoffman, L. M. & Carpenter, M. K. Characterization and culture of human embryonic stem cells. Nature Biotech. 23, 699–708 (2005).

    CAS  Google Scholar 

  181. Xu, C. Characterization and evaluation of human embryonic stem cells. Methods Enzymol. 420, 18–37 (2006).

    CAS  PubMed  Google Scholar 

  182. Klimanskaya, I. & McMahon, J. in Handbook of Stem Cells Vol. 1 Ch. 41 (eds Lanza, R. et al.) 437–449 (Academic, Amsterdam, 2004).

    Google Scholar 

  183. The International Stem CellInitiative et al. Characterization of human embryonic stem cell lines by the International Stem Cell Initiative. Nature Biotech. 25, 803–816 (2007).

  184. Menendez, P., Wang, L. & Bhatia, M. Genetic manipulation of human embryonic stem cells: a system to study early human development and potential therapeutic applications. Curr. Gene Ther. 5, 375–385 (2005).

    CAS  PubMed  Google Scholar 

  185. Gropp, M. & Reubinoff, B. Lentiviral vector-mediated gene delivery into human embryonic stem cells. Methods Enzymol. 420, 64–81 (2006).

    CAS  PubMed  Google Scholar 

  186. Draper, J. S. et al. Recurrent gain of chromosomes 17q and 12 in cultured human embryonic stem cells. Nature Biotech. 22, 53–54 (2004).

    CAS  Google Scholar 

  187. Gertow, K. et al. Trisomy 12 in HESC leads to no selective in vivo growth advantage in teratomas, but induces an increased abundance of renal development. J. Cell Biochem. 100, 1518–1525 (2007).

    CAS  PubMed  Google Scholar 

  188. Mitalipova, M. M. et al. Preserving the genetic integrity of human embryonic stem cells. Nature Biotech. 23, 19–20 (2005).

    CAS  Google Scholar 

  189. Zwaka, T. P. & Thomson, J. A. Homologous recombination in human embryonic stem cells. Nature Biotech. 21, 319–321 (2003).

    CAS  Google Scholar 

  190. Cowan, C. A. et al. Derivation of embryonic stem-cell lines from human blastocysts. N. Engl. J. Med. 350, 1353–1356 (2004).

    CAS  PubMed  Google Scholar 

  191. Byrne, J. A. et al. Producing primate embryonic stem cells by somatic cell nuclear transfer. Nature 450, 497–502 (2007).

    CAS  PubMed  Google Scholar 

  192. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 20 Nov 2007 (doi:10.1016/j.cell.2007.11.019).

    CAS  PubMed  Google Scholar 

  193. Yu, J. et al. Induced pluripotent stem cell lines derived from human somatic cells. Science 20 Nov 2007 (doi:10.1126/science.1151526).

    CAS  PubMed  Google Scholar 

  194. Meng, X. et al. Endometrial regenerative cells: a novel stem cell population. J. Trans. Med. 15 Nov 2007 (doi:10.1186/1479-5876-5-57).

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Irina Klimanskaya.

Ethics declarations

Competing interests

I.K. and R.L. are employees of Advanced Cell Technologies, a biotechnology company in the field of stem cells and regenerative medicine.

Related links

Related links

FURTHER INFORMATION

Advanced Cell Technology

European Molecular Biology Laboratory

NIH Stem Cell Information

UK Stem Cell Bank

Glossary

Pluripotent

In cell biology this term is usually used to indicate that pluripotent cells can develop into derivatives of all three germ layers.

Organ-derived stem cells

These are often called 'adult' or 'somatic' stem cells; adult in this case meaning 'differentiated' or 'other than embryonic', because multipotent stem cells have also been isolated from extraembryonic and perinatal tissues.

Feeder cells

Cells of a different type and often different species that are used in a co-culture system to help maintain embryonic stem cells (ESCs) undifferentiated and mitotically inactivated to prevent overgrowing. Traditionally mouse embryonic fibroblasts were used as feeders for mouse and human ESCs, but in anticipation of using human ESC derivatives in the clinic, novel human ESC culture systems have been developed that use human cell lines as feeders or no feeder cells at all.

Teratoma

A rare tumour type that typically arises in the gonads and demonstrates mixed cellular populations of all three embryonic germ layers. Investigators can assess the differentiation capacity of stem cells by injection of pluripotent cells into laboratory animals and inducing the formation of teratomas in situ.

Bromodeoxyuridine

(BrdU). 5-bromo-2-deoxyuridine is a synthetic analogue of the nucleoside thymidine. BrdU is used to label proliferating cells within a given time-frame in vitro or in vivo. It incorporates into the newly replicated DNA strands and can be detected with anti-BrdU antibodies.

Blastocyst

A blastocyst is a multi-cell structure formed by a developing mammalian embryo at the early stages of development that looks like a spheroid formed by an outer cell layer — trophectoderm, a cavity — a blastocoel and an inner cell mass (ICM). In further development, a trophoblast gives rise to extraembryonic tissues, while the ICM develops into a new organism. Cells of the ICM are pluripotent and can also produce embryonic stem cells.

Parthenote

Parthenote, parthenogenesis is derived from the Greek 'parthenos', which means virgin. In this article, it pertains to activated unfertilized oocytes that are capable of undergoing the cleavage division and forming blastocysts. It is accepted that parthenote embryos in mammals are not capable of forming extraembryonic tissues and thus cannot complete normal development.

Reprogramming

Occurs naturally in regenerative organisms (dedifferentiation). Induced experimentally in mammalian cells by nuclear transfer, cell fusion or genetic manipulation of in vitro culture.

Mulitpotent

Can form multiple lineages that can give rise to several kinds of cells, tissues or structures, for example, haematopoietic stem cells.

Cybrid

Is a cell produced by the fusing of a eukaryotic cell with an enucleated cell or a cytoplast.

Totipotent

Sufficient to form an entire organism. Totipotency is seen in zygote and plant meristem cells, but has not demonstrated for any vertebrate stem cell.

Morula

A stage of early embryonic development when an embryo consists of a cluster of cells.

C-peptide

C-peptide is a product of proinsulin when it is split, producing insulin and C-peptide after release from the pancreas.

Embryoid bodies

Spherical cell clusters observed after spontaneous or induced differentiation of embryonic stem cells in culture. Embryoid bodies show differentiation that recapitulates the early stages of mammalian embryonic development, including cell types derived from endoderm, mesoderm and ectodermal lineages.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Klimanskaya, I., Rosenthal, N. & Lanza, R. Derive and conquer: sourcing and differentiating stem cells for therapeutic applications. Nat Rev Drug Discov 7, 131–142 (2008). https://doi.org/10.1038/nrd2403

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd2403

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