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.

Generation of transgenic non-human primates with germline transmission

Abstract

The common marmoset (Callithrix jacchus) is increasingly attractive for use as a non-human primate animal model in biomedical research. It has a relatively high reproduction rate for a primate, making it potentially suitable for transgenic modification. Although several attempts have been made to produce non-human transgenic primates, transgene expression in the somatic tissues of live infants has not been demonstrated by objective analyses such as polymerase chain reaction with reverse transcription or western blots. Here we show that the injection of a self-inactivating lentiviral vector in sucrose solution into marmoset embryos results in transgenic common marmosets that expressed the transgene in several organs. Notably, we achieved germline transmission of the transgene, and the transgenic offspring developed normally. The successful creation of transgenic marmosets provides a new animal model for human disease that has the great advantage of a close genetic relationship with humans. This model will be valuable to many fields of biomedical research.

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: Self-inactivating lentiviral vector-derived EGFP transgenic marmosets.
Figure 2: Transgene insertions in several infant tissues.
Figure 3: Transgene transcription and expression in several infant tissues.
Figure 4: Germline transmission of the transgene.

References

  1. Mansfield, K. Marmoset models commonly used in biomedical research. Comp. Med. 53, 383–392 (2003)

    CAS  PubMed  Google Scholar 

  2. Chan, A. W., Homan, E. J., Ballou, L. U., Burns, J. C. & Bremel, R. D. Transgenic cattle produced by reverse-transcribed gene transfer in oocytes. Proc. Natl Acad. Sci. USA 95, 14028–14033 (1998)

    Article  ADS  CAS  Google Scholar 

  3. Hofmann, A. et al. Efficient transgenesis in farm animals by lentiviral vectors. EMBO Rep. 4, 1054–1060 (2003)

    Article  CAS  Google Scholar 

  4. Hofmann, A. et al. Generation of transgenic cattle by lentiviral gene transfer into oocytes. Biol. Reprod. 71, 405–409 (2004)

    Article  CAS  Google Scholar 

  5. Chan, A. W., Chong, K. Y., Martinovich, C., Simerly, C. & Schatten, G. Transgenic monkeys produced by retroviral gene transfer into mature oocytes. Science 291, 309–312 (2001)

    Article  ADS  CAS  Google Scholar 

  6. Wolfgang, M. J. et al. Rhesus monkey placental transgene expression after lentiviral gene transfer into preimplantation embryos. Proc. Natl Acad. Sci. USA 98, 10728–10732 (2001)

    Article  ADS  CAS  Google Scholar 

  7. Yang, S. H. et al. Towards a transgenic model of Huntington’s disease in a non-human primate. Nature 453, 921–924 (2008)

    Article  ADS  CAS  Google Scholar 

  8. Rogers, C. S. et al. Disruption of the CFTR gene produces a model of cystic fibrosis in newborn pigs. Science 321, 1837–1841 (2008)

    Article  ADS  CAS  Google Scholar 

  9. Rogers, C. S. et al. Production of CFTR-null and CFTR-ΔF508 heterozygous pigs by adeno-associated virus-mediated gene targeting and somatic cell nuclear transfer. J. Clin. Invest. 118, 1571–1577 (2008)

    Article  ADS  CAS  Google Scholar 

  10. Miyoshi, H., Blomer, U., Takahashi, M., Gage, F. H. & Verma, I. M. Development of a self-inactivating lentivirus vector. J. Virol. 72, 8150–8157 (1998)

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Ross, C. N., French, J. A. & Orti, G. Germ-line chimerism and paternal care in marmosets (Callithrix kuhlii). Proc. Natl Acad. Sci. USA 104, 6278–6282 (2007)

    Article  ADS  CAS  Google Scholar 

  12. 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 

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

    Article  ADS  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  15. Nakagawa, M. et al. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nature Biotechnol. 26, 101–106 (2008)

    Article  CAS  Google Scholar 

  16. Iwanami, A. et al. Transplantation of human neural stem cells for spinal cord injury in primates. J. Neurosci. Res. 80, 182–190 (2005)

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  18. Iwanami, A. et al. Establishment of graded spinal cord injury model in a nonhuman primate: the common marmoset. J. Neurosci. Res. 80, 172–181 (2005)

    Article  CAS  Google Scholar 

  19. Eslamboli, A. Marmoset monkey models of Parkinson’s disease: which model, when and why? Brain Res. Bull. 68, 140–149 (2005)

    Article  Google Scholar 

  20. Ando, K. et al. Neurobehavioral protection by single dose l-deprenyl against MPTP-induced parkinsonism in common marmosets. Psychopharmacology (Berl.) 195, 509–516 (2008)

    Article  CAS  Google Scholar 

  21. Sasaki, E. et al. Establishment of novel embryonic stem cell lines derived from the common marmoset (Callithrix jacchus). Stem Cells 23, 1304–1313 (2005)

    Article  CAS  Google Scholar 

  22. Lopata, A., Summers, P. M. & Hearn, J. P. Births following the transfer of cultured embryos obtained by in vitro and in vivo fertilization in the marmoset monkey (Callithrix jacchus). Fertil. Steril. 50, 503–509 (1988)

    Article  CAS  Google Scholar 

  23. Summers, P. M., Shephard, A. M., Taylor, C. T. & Hearn, J. P. The effects of cryopreservation and transfer on embryonic development in the common marmoset monkey, Callithrix jacchus . J. Reprod. Fertil. 79, 241–250 (1987)

    Article  CAS  Google Scholar 

  24. Kuederling, I., Schneiders, A., Sonksen, J., Nayudu, P. L. & Hodges, J. K. Non-invasive collection of ejaculates from the common marmoset (Callithrix jacchus) using penile vibrostimulation. Am. J. Primatol. 52, 149–154 (2000)

    Article  CAS  Google Scholar 

  25. Summers, P. M., Shephard, A. M., Taylor, C. T. & Hearn, J. P. The effects of cryopreservation and transfer on embryonic development in the common marmoset monkey, Callithrix jacchus . J. Reprod. Fertil. 79, 241–250 (1987)

    Article  CAS  Google Scholar 

  26. Bai, Y. et al. Effective transduction and stable transgene expression in human blood cells by a third-generation lentiviral vector. Gene Ther. 10, 1446–1457 (2003)

    Article  CAS  Google Scholar 

  27. Ito, R. et al. Novel monoclonal antibodies recognizing different subsets of lymphocytes from the common marmoset (Callithrix jacchus). Immunol. Lett. 121, 116–122 (2008)

    Article  ADS  CAS  Google Scholar 

Download references

Acknowledgements

We thank F. Toyoda, S. Ohba, T. Inoue, Y. Sawada and M. Yokoyama for technical assistance with the animal experiments and care. E.S. is an associate professor of the Global COE program for human metabolomic systems biology assigned to Keio University. This study was also supported by the Global COE program for Education and Research Centre for Stem Cell Medicine from the Ministry of Education, Culture, Sports, Science and Technology (MEXT), the Japanese Government to Keio University. This study was also supported by funds from Solution-Oriented Research for Science and Technology (SORST) of the Japan Science and Technology Agency and grants from MEXT to H.O. and from Special Coordination Funds for Promoting Science and Technology of MEXT to S.H.

Author Contributions E.S. designed the experiments, conducted the project, and wrote the paper. A.S., Y.S., T.E., I.T. and R.H. assisted in embryological technique development. K.H., R.O. and M.K. developed surgical techniques for embryo collection and transfer. H.S., C.K. and C.Y. performed or assisted with the real-time PCR and parentage evaluation test. S.S. and T.M. assisted with the Southern blot analysis and tissue collection. M.I. raised the anti-marmoset CD45 antibody. R.I. performed the FACS analysis, and K.K. performed the immunohistochemical analysis. H.M. provided the lentiviral vectors. Y.T., H.O., S.H., N.T. and T.N. designed the project, and H.O., S.H. and N.T. also participated in writing the paper. The whole project was supervised by E.S. and H.O.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Erika Sasaki or Hideyuki Okano.

Supplementary information

Supplementary Information

This file contains Supplementary Tables 1-3 and Supplementary Figures 1-5 with Legends. (PDF 1759 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sasaki, E., Suemizu, H., Shimada, A. et al. Generation of transgenic non-human primates with germline transmission. Nature 459, 523–527 (2009). https://doi.org/10.1038/nature08090

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature08090

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