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.

  • Letter
  • Published:

Human oocytes reprogram adult somatic nuclei of a type 1 diabetic to diploid pluripotent stem cells

Subjects

Abstract

The transfer of somatic cell nuclei into oocytes can give rise to pluripotent stem cells that are consistently equivalent to embryonic stem cells1,2,3, holding promise for autologous cell replacement therapy4,5. Although methods to induce pluripotent stem cells from somatic cells by transcription factors6 are widely used in basic research, numerous differences between induced pluripotent stem cells and embryonic stem cells have been reported7,8,9,10,11, potentially affecting their clinical use. Because of the therapeutic potential of diploid embryonic stem-cell lines derived from adult cells of diseased human subjects, we have systematically investigated the parameters affecting efficiency of blastocyst development and stem-cell derivation. Here we show that improvements to the oocyte activation protocol, including the use of both kinase and translation inhibitors, and cell culture in the presence of histone deacetylase inhibitors, promote development to the blastocyst stage. Developmental efficiency varied between oocyte donors, and was inversely related to the number of days of hormonal stimulation required for oocyte maturation, whereas the daily dose of gonadotropin or the total number of metaphase II oocytes retrieved did not affect developmental outcome. Because the use of concentrated Sendai virus for cell fusion induced an increase in intracellular calcium concentration, causing premature oocyte activation, we used diluted Sendai virus in calcium-free medium. Using this modified nuclear transfer protocol, we derived diploid pluripotent stem-cell lines from somatic cells of a newborn and, for the first time, an adult, a female with type 1 diabetes.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Developmental potential of somatic cell nuclear transfer oocytes.
Figure 2: Chromosome condensation and spindle assembly after somatic cell nuclear transfer.
Figure 3: Derivation of diploid NT-ES cells from adult somatic cells.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

Microarray data are available at GEO under accession numbers GSE54849 and GSE54876.

References

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

    Article  CAS  ADS  Google Scholar 

  2. 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)

    Article  CAS  ADS  Google Scholar 

  3. Wakayama, S. et al. Equivalency of nuclear transfer-derived embryonic stem cells to those derived from fertilized mouse blastocysts. Stem Cells 24, 2023–2033 (2006)

    Article  CAS  Google Scholar 

  4. Tabar, V. et al. Therapeutic cloning in individual parkinsonian mice. Nature Med. 14, 379–381 (2008)

    Article  CAS  Google Scholar 

  5. Rideout, W. M., III, Hochedlinger, K., Kyba, M., Daley, G. Q. & Jaenisch, R. Correction of a genetic defect by nuclear transplantation and combined cell and gene therapy. Cell 109, 17–27 (2002)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  7. Koyanagi-Aoi, M. et al. Differentiation-defective phenotypes revealed by large-scale analyses of human pluripotent stem cells. Proc. Natl Acad. Sci. USA 110, 20569–20574 (2013)

    Article  CAS  ADS  Google Scholar 

  8. Wang, T. et al. Subtelomeric hotspots of aberrant 5-hydroxymethylcytosine-mediated epigenetic modifications during reprogramming to pluripotency. Nature Cell Biol. 15, 700–711 (2013)

    Article  CAS  Google Scholar 

  9. Ohi, Y. et al. Incomplete DNA methylation underlies a transcriptional memory of somatic cells in human iPS cells. Nature Cell Biol. 13, 541–549 (2011)

    Article  CAS  Google Scholar 

  10. Ruiz, S. et al. Identification of a specific reprogramming-associated epigenetic signature in human induced pluripotent stem cells. Proc. Natl Acad. Sci. USA 109, 16196–16201 (2012)

    Article  CAS  ADS  Google Scholar 

  11. Pick, M. et al. Clone- and gene-specific aberrations of parental imprinting in human induced pluripotent stem cells. Stem Cells 27, 2686–2690 (2009)

    Article  CAS  Google Scholar 

  12. Noggle, S. et al. Human oocytes reprogram somatic cells to a pluripotent state. Nature 478, 70–75 (2011)

    Article  CAS  ADS  Google Scholar 

  13. Van Thuan, N. et al. The histone deacetylase inhibitor scriptaid enhances nascent mRNA production and rescues full-term development in cloned inbred mice. Reproduction 138, 309–317 (2009)

    Article  CAS  Google Scholar 

  14. Paull, D. et al. Nuclear genome transfer in human oocytes eliminates mitochondrial DNA variants. Nature 493, 632–637 (2013)

    Article  CAS  ADS  Google Scholar 

  15. Winston, N. J. Stability of cyclin B protein during meiotic maturation and the first mitotic cell division in mouse oocytes. Biol. Cell 89, 211–219 (1997)

    Article  CAS  Google Scholar 

  16. Stern, S., Rayyis, A. & Kennedy, J. F. Incorporation of amino acids during maturation in vitro by the mouse oocyte: effect of puromycin on protein synthesis. Biol. Reprod. 7, 341–346 (1972)

    Article  CAS  Google Scholar 

  17. Braude, P., Bolton, V. & Moore, S. Human gene expression first occurs between the four- and eight-cell stages of preimplantation development. Nature 332, 459–461 (1988)

    Article  CAS  ADS  Google Scholar 

  18. Tachibana, M. et al. Human embryonic stem cells derived by somatic cell nuclear transfer. Cell 153, 1228–1238 (2013)

    Article  CAS  Google Scholar 

  19. Fan, Y. et al. Derivation of cloned human blastocysts by histone deacetylase inhibitor treatment after somatic cell nuclear transfer with beta-thalassemia fibroblasts. Stem Cells Dev. 20, 1951–1959 (2011)

    Article  CAS  Google Scholar 

  20. Wakayama, T., Perry, A. C., Zuccotti, M., Johnson, K. R. & Yanagimachi, R. Full-term development of mice from enucleated oocytes injected with cumulus cell nuclei. Nature 394, 369–374 (1998)

    Article  CAS  ADS  Google Scholar 

  21. Liu, L., Oldenbourg, R., Trimarchi, J. R. & Keefe, D. L. A reliable, noninvasive technique for spindle imaging and enucleation of mammalian oocytes. Nature Biotechnol. 18, 223–225 (2000)

    Article  CAS  Google Scholar 

  22. Gassmann, R. et al. Borealin: a novel chromosomal passenger required for stability of the bipolar mitotic spindle. J. Cell Biol. 166, 179–191 (2004)

    Article  CAS  Google Scholar 

  23. Cohen, M. A., Lindheim, S. R. & Sauer, M. V. Donor age is paramount to success in oocyte donation. Hum. Reprod. 14, 2755–2758 (1999)

    Article  CAS  Google Scholar 

  24. Bar-Hava, I. et al. Controlled ovarian hyperstimulation: does prolonged stimulation justify cancellation of in vitro fertilization cycles? Gynecol. Endocrinol. 21, 232–234 (2005)

    Article  Google Scholar 

  25. Lister, R. et al. Hotspots of aberrant epigenomic reprogramming in human induced pluripotent stem cells. Nature 471, 68–73 (2011)

    Article  CAS  ADS  Google Scholar 

  26. Gore, A. et al. Somatic coding mutations in human induced pluripotent stem cells. Nature 471, 63–67 (2011)

    Article  CAS  ADS  Google Scholar 

  27. Egli, D. et al. Reprogramming within hours following nuclear transfer into mouse but not human zygotes. Nature Commun. 2, 488 (2011)

    Article  ADS  Google Scholar 

  28. Egli, D. & Chia, G. A protocol for embryonic stem cell derivation by somatic cell nuclear transfer into human oocytes. Protocol Exch http://dx.doi.org/10.1038/protex.2014.013 (2014)

  29. Klitzman, R. & Sauer, M. V. Payment of egg donors in stem cell research in the USA. Reprod. Biomed. Online 18, 603–608 (2009)

    Article  Google Scholar 

  30. Egli, D. et al. Impracticality of egg donor recruitment in the absence of compensation. Cell Stem Cell 9, 293–294 (2011)

    Article  CAS  Google Scholar 

  31. Choudhary, M. et al. Egg sharing for research: a successful outcome for patients and researchers. Cell Stem Cell 10, 239–240 (2012)

    Article  CAS  Google Scholar 

  32. The Ethics Committee of the American Society for Reproductive Medicine Financial compensation of oocyte donors. Fertil. Steril. 88, 305–309 (2007)

  33. Daley, G. Q. et al. Ethics. The ISSCR guidelines for human embryonic stem cell research. Science 315, 603–604 (2007)

    Article  CAS  Google Scholar 

  34. Tachibana, M., Sparman, M. & Mitalipov, S. Chromosome transfer in mature oocytes. Nature Protocols 5, 1138–1147 (2010)

    Article  CAS  Google Scholar 

  35. Egli, D. & Eggan, K. Nuclear transfer into mouse oocytes. J. Vis. Exp. 30, 116 (2006)

    Google Scholar 

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

    Article  CAS  Google Scholar 

  37. Hua, H. et al. iPSC-derived beta cells model diabetes due to glucokinase deficiency. J. Clin. Invest. 123, 3146–3153 (2013)

    Article  CAS  Google Scholar 

  38. Shang, L. et al. Beta cell dysfunction due to increased ER stress in a stem cell model of Wolfram syndrome. Diabetes 63, 923–933 (2014

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This research was supported the New York Stem Cell Foundation (NYSCF) and a New York State Stem Cell Science (NYSTEM) IIRP Award no. C026184, and the Russell Berrie Foundation Program in Cellular Therapies of Diabetes. We thank S. Mitalipov for helpful discussions and providing reagents, S. Micucci for counting cells in S-phase, and Z. Hall for critical reading of the manuscript. D.E. is a NYSCF-Robertson Investigator.

Author information

Authors and Affiliations

Authors

Contributions

M.V.S. supervised the research oocyte donation program and retrieved oocytes. D.E. designed, performed and interpreted nuclear transfer experiments, derived ES cells with M.Y., and wrote the paper with input from all authors. M.Y. performed statistical analysis, M.Y. and B.J. performed stem cell characterization and differentiation, L.C.B. performed neuronal differentiation, I.S. and N.B. performed gene expression analysis, M.W.N. assisted with calcium experiments, D.H.K. performed data analysis, D.P. assisted in nuclear transfer experiments, R.W.P. collected developmental data of IVF embryos, M.F. made the skin biopsy, E.G. coordinated human subjects research, R.S.G. wrote the IRB protocol, R.L.L. contributed project planning, S.L.S. created the environment specifically for this work.

Corresponding author

Correspondence to Dieter Egli.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Efficiency of parthenogenetic development beyond the cleavage stage.

Shown is the percentage of oocytes giving rise to stem cell lines, blastocysts but no stem cell lines, and morulae as the percentage of the number of oocytes cleaved. Data are from manuscript references12,14 and displayed here in a direct comparison. The number of oocytes used is indicated above the column. The number of repeats, with oocytes from different donors, is indicated in parenthesis. Statistical analysis using Chi-square test was performed by comparing the total number of cells formed in each condition. Morulae were assigned 15 cells, blastocysts or blastocysts that gave rise to stem-cell lines 30 cells, reflecting the estimated minimal cell count for each group.

Extended Data Figure 2 Development to the blastocyst stage and transcriptional activation of the transferred genome.

a, Blastocyst derived after nuclear transfer of a BJ fibroblast genome (neonatal foreskin fibroblasts). b, Blastocysts derived after nuclear transfer of an adult skin fibroblast genome. c, Three different nuclear transfer ES cell outgrowths. Time post blastocyst plating is indicated. Scale bar, 10 µm.

Extended Data Figure 3 Fluorescence imaging with the calcium-responsive dye Fluo-4.

a, Human oocytes were incubated in medium containing Fluo-4 for 30 min, imaged for fluorescence, and concentrated Sendai virus was added below the plasma membrane. Shown are two oocytes for each condition or time point. Note that in the absence of calcium in the medium, fluorescence did not increase, whereas a small increase in fluorescence seems to occur in calcium-containing medium. Time point after addition of the virus is indicated. b, Incubation of a human oocyte in 3 µM of the calcium ionophore ionomycin as a positive control. Scale bar, 10 µm.

Extended Data Figure 4 Somatic cell nuclear transfer in the absence of calcium.

a, Immunochemistry to determine chromosome condensation and histone phosphorylation after transfer of a somatic cell at interphase. Scale bar, 5 µm. b, High-quality blastocysts obtained after nuclear transfer with the manipulations conducted in the absence of calcium. Scale bar, 10 µm.

Extended Data Figure 5 Effect of FBS on blastocyst morphology and ES cell derivation.

a, Blastocyst generated by somatic cell nuclear transfer in the presence or absence of FBS. b, Inner cell mass 6 days post plating. Arrows point to laser marks used to ablate the remaining trophectoderm cells. Scale bar, 20 µm.

Extended Data Figure 6 Characterization of NT-ES cells from male foreskin BJ fibroblasts.

ac, Karyotypes and pluripotency marker expression in three NT-ES cell lines derived from male foreskin BJ fibroblasts. The somatic donor cell used for transfer carries a GFP transgene. Scale bar, 50 µm.

Extended Data Figure 7 Differentiation of NT-ES cell lines made from male foreskin BJ fibroblasts.

Embryoid bodies and teratomas are shown. Scale bar, 50 µm.

Extended Data Figure 8 Retrospective analysis of the developmental potential of nuclear transfer oocytes.

Shown is the percentage of oocytes developing beyond the cleavage stage, as percentage of eggs progressing beyond the 1-cell stage. Because oocytes of a donor were used to compare two different conditions, if for a particular comparison the added number of oocyte donors exceeds 18, it indicates that these conditions were tested in parallel using oocytes of the same donor. The total number of oocytes used for analysis remained constant. a, Average of the 154 oocytes of 18 donors. The total number of 154 oocytes is not equal to the number of oocytes donated by the 18 donors, but is the number of oocytes used for the study of developmental potential after somatic cell nuclear transfer. b, Analysis with regard to factors relevant to the hormonal treatment of oocyte donor. c, Factors relevant to the manipulation. *Blastocysts generated without caffeine treatment also contained no FBS in the culture medium. Because of the effect of FBS on inner cell mass morphology (Extended Data Fig. 5), FBS is likely the more relevant factor. d, Analysis regarding cell source and use of FBS for culture. n.s., non significant. Statistical analysis using Chi-square test was performed by comparing the total number of cells formed in each condition. Morulae were assigned 15 cells, blastocysts or blastocysts that gave rise to stem-cell lines 30 cells, reflecting the estimated minimal cell count for each group.

Extended Data Table 1 Complete list of samples used in global gene expression analyses
Extended Data Table 2 Short tandem repeat (STR) profiles of nuclear transfer ES cell lines

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yamada, M., Johannesson, B., Sagi, I. et al. Human oocytes reprogram adult somatic nuclei of a type 1 diabetic to diploid pluripotent stem cells. Nature 510, 533–536 (2014). https://doi.org/10.1038/nature13287

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature13287

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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