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:

Peptide nanofibrils boost retroviral gene transfer and provide a rapid means for concentrating viruses

Abstract

Inefficient gene transfer and low virion concentrations are common limitations of retroviral transduction1. We and others have previously shown that peptides derived from human semen form amyloid fibrils that boost retroviral gene delivery by promoting virion attachment to the target cells2,3,4,5,6,7,8. However, application of these natural fibril-forming peptides is limited by moderate efficiencies, the high costs of peptide synthesis, and variability in fibril size and formation kinetics. Here, we report the development of nanofibrils that self-assemble in aqueous solution from a 12-residue peptide, termed enhancing factor C (EF-C). These artificial nanofibrils enhance retroviral gene transfer substantially more efficiently than semen-derived fibrils or other transduction enhancers. Moreover, EF-C nanofibrils allow the concentration of retroviral vectors by conventional low-speed centrifugation, and are safe and effective, as assessed in an ex vivo gene transfer study. Our results show that EF-C fibrils comprise a highly versatile, convenient and broadly applicable nanomaterial that holds the potential to significantly facilitate retroviral gene transfer in basic research and clinical applications.

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: Peptides derived from the HIV-1 glycoprotein gp120 enhance virus infection.
Figure 2: Structural characterization and molecular modelling of EF-C fibrils.
Figure 3: EF-C fibrils bind, precipitate and concentrate virions.
Figure 4: EF-C fibrils are broad-based transduction enhancers.
Figure 5: EF-C fibrils can be immobilized (a–c) and allow efficient gene transfer into mice (d–f).

Similar content being viewed by others

References

  1. Mátrai, J., Chuah, M. K. & VandenDriessche, T. Recent advances in lentiviral vector development and applications. Mol. Ther. 18, 477–490 (2010).

    Article  Google Scholar 

  2. Münch, J. et al. Semen-derived amyloid fibrils drastically enhance HIV infection. Cell 131, 1059–1071 (2007).

    Article  Google Scholar 

  3. Roan, N. R. et al. The cationic properties of SEVI underlie its ability to markedly enhance HIV infection. J. Virol. 83, 73–80 (2009).

    Article  CAS  Google Scholar 

  4. Kim, K. et al. Semen-mediated enhancement of HIV infection is donor-dependent and correlates with the levels of SEVI. Retrovirology 7, 55 (2010).

    Article  Google Scholar 

  5. Wurm, M. et al. The influence of semen-derived enhancer of virus infection on the efficiency of retroviral gene transfer. J. Gene Med. 12, 137–146 (2010).

    CAS  Google Scholar 

  6. Wurm, M. et al. Improved lentiviral gene transfer into human embryonic stem cells grown in co-culture with murine feeder and stroma cells. Biol. Chem. 392, 887–895 (2011).

    Article  CAS  Google Scholar 

  7. Roan, N. R. et al. Peptides released by physiological cleavage of semen coagulum proteins form amyloids that enhance HIV infection. Cell Host Microbe 15, 541–550 (2011).

    Article  Google Scholar 

  8. Arnold, F. et al. Naturally occurring fragments from two distinct regions of the prostatic acid phosphatase form amyloidogenic enhancers of HIV infection. J. Virol. 86, 1244–1249 (2012).

    Article  CAS  Google Scholar 

  9. Kwong, P. D. et al. Structure of an HIV gp120 envelope glycoprotein in complex with the CD4 receptor and a neutralizing human antibody. Nature 393, 648–659 (1998).

    Article  CAS  Google Scholar 

  10. Knowles, T. P. & Buehler, M. J. Nanomechanics of functional and pathological amyloid materials. Nature Nanotech. 6, 469–479 (2011).

    Article  CAS  Google Scholar 

  11. Dalhaimer, P., Bates, F. S. & Discher, D. E. Single molecule visualization of stable, stiffness-tunable, flow-conforming worm micelles. Macromolecules 36, 6873–6877 (2003).

    Article  CAS  Google Scholar 

  12. Cerf, E. et al. Antiparallel β-sheet: a signature structure of the oligomeric amyloid β-peptide. Biochem. J. 421, 415–423 (2009).

    Article  CAS  Google Scholar 

  13. Makin, O. S. & Serpell, L. C. Structures for amyloid fibrils. FEBS J. 272, 5950–5961 (2005).

    Article  CAS  Google Scholar 

  14. Shaytan, A. K. et al. Self-assembling nanofibers from thiophene–peptide diblock oligomers: a combined experimental and computer simulations study. ACS Nano 5, 6894–6909 (2011).

    Article  CAS  Google Scholar 

  15. Tiscornia, G., Singer, O. & Verma, I. M. Production and purification of lentiviral vectors. Nature Protoc. 1, 241–245 (2006).

    Article  CAS  Google Scholar 

  16. Sandrin, V. et al. Lentiviral vectors pseudotyped with a modified RD114 envelope glycoprotein show increased stability in sera and augmented transduction of primary lymphocytes an CD34+ cells derived from human and nonhuman primates. Blood 100, 823–832 (2002).

    Article  CAS  Google Scholar 

  17. Schambach, A. et al. Equal potency of gammaretroviral and lentiviral SIN vectors for expression of O6-methylguanine-DNA methyltransferase in hematopoietic cells. Mol. Ther. 13, 391–400 (2006).

    Article  CAS  Google Scholar 

  18. Schambach, A. et al. Overcoming promoter competition in packaging cells improves production of self-inactivating retroviral vectors. Gene Ther. 13, 1524–1533 (2006).

    Article  CAS  Google Scholar 

  19. Dull, T. et al. A third-generation lentivirus vector with a conditional packaging system. J. Virol. 72, 8463–8471 (1998).

    CAS  Google Scholar 

  20. Koeffler, H. P. & Golde, D. W. Human myeloid leukemia cell lines: a review. Blood 56, 344–350 (1980).

    CAS  Google Scholar 

  21. Leyva, F. J., Anzinger, J. J., McCoy, J. P. & Kruth, H. S. Evaluation of transduction efficiency in macrophage colony-stimulating factor differentiated human macrophages using HIV-1 based lentiviral vectors. BMC Biotechnol. 11, 13 (2011).

    Article  CAS  Google Scholar 

  22. Manning, J. S., Hackett, A. J. & Darby, N. B. Effect of polycations on sensitivity of BALD-3T3 cells to murine leukemia and sarcoma virus infectivity. Appl. Microbiol. 22, 1162–1163 (1971).

    CAS  Google Scholar 

  23. Cornetta, K. & Anderson, W. F. Protamine sulfate as an effective alternative to polybrene in retroviral-mediated genetransfer: implications for human gene therapy. J. Virol. Methods 23, 187–194 (1989).

    Article  CAS  Google Scholar 

  24. Kaplan, M. M., Wiktor, T. J., Maes, R. F., Campbell, J. B. & Koprowski, H. Effect of polyions on the infectivity of rabies virus in tissue culture: construction of a single-cycle growth curve. J. Virol. 1, 145–151 (1967).

    CAS  Google Scholar 

  25. Hanenberg, H. et al. Colocalization of retrovirus and target cells on specific fibronectin fragments increases genetic transduction of mammalian cells. Nature Med. 2, 876–882 (1996).

    Article  CAS  Google Scholar 

  26. Hanenberg, H. et al. Optimization of fibronectin-assisted retroviral gene transfer into human CD34+ hematopoietic cells. Hum. Gene Ther. 8, 2193–2206 (1997).

    Article  CAS  Google Scholar 

  27. Pollok, K. E. et al. High efficiency gene transfer into normal and adenosine deaminase deficient T lymphocytes is mediated by transduction on recombinant fibronectin fragments. J. Virol. 72, 4882–4892 (1998).

    CAS  Google Scholar 

  28. Millington, M., Arndt, A., Boyd, M., Applegate, T. & Shen, S. Towards a clinically relevant lentiviral transduction protocol for primary human CD34 hematopoietic stem/progenitor cells. PLoS ONE 4, e6461 (2009).

    Article  Google Scholar 

  29. Kustikova, O. S. et al. Retroviral vector insertion sites associated with dominant hematopoietic clones mark ‘stemness’ pathways. Blood 109, 1897–1907 (2007).

    Article  CAS  Google Scholar 

  30. Knowles, T. P. et al. Role of intermolecular forces in defining material properties of protein nanofibrils. Science 318, 1900–1903 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank S. Liu for support in the initial phase of the project and D. Krnavek and M. Schlaier for expert technical assistance. The authors also thank W. Mothes for providing the MLV GAG-CFP plasmid, B. Böhm for Bon cells, J. von Einem for HFF cells, the AIDS Research and Reference Program for U87-MG and TZM-bl cells, and N. Landau for CEMx-M7 (CEMx174 5.25 M7) cells. This work was supported by a grant from the German Research Foundation to J.M. Molecular simulations were performed using the resources of Moscow University Supercomputing Center.

Author information

Authors and Affiliations

Authors

Contributions

M.Y. generated virus stocks and performed most transductions. C.M. and T.W. assisted in writing the manuscript and were responsible for the structural elucidation of EF-C fibrils. A.K.S., P.G.K. and A.R.K. performed molecular simulations. V.V. and H.G. conducted the ex vivo gene transfer study. C.W.B. and X.S. performed CD analysis. F.A. and D.P. performed coating experiments and measured zeta potentials. O.Z. and J.M. performed Congo Red and ThT assays, S.M.U. was responsible for the analysis of immobilized EF-C by confocal microscopy. D.S. and C.G. performed the fusion assay and flow cytometry assays. P.W. assisted in microscopy. O.L. and T.S. studied the EF-C interaction with virions and cells. J.B. provided retro- and lentiviral vectors and knowhow. H.S. and K.S. isolated and provided human stem cells and performed colony-forming unit assays. L.S. and W.G.F. synthesized and provided peptides. N.R.R. assisted in planning and writing. T.K. analysed AFM data. F.K. and J.M. planned research, analysed data and wrote the manuscript. All authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Jan Münch.

Ethics declarations

Competing interests

M. Yo., F. Ki. and J. Mü. filed for a patent to use EF-C fibrils as tranduction and infection enhancer.

Supplementary information

Supplementary information (PDF 2826 kb)

Supplementary movie S1

Supplementary movie S1 (MOV 17643 kb)

Supplementary movie S2

Supplementary movie S2 (MOV 10073 kb)

Supplementary movie S3

Supplementary movie S3 (AVI 1596 kb)

Supplementary movie S4

Supplementary movie S4 (AVI 1097 kb)

Supplementary movie S5

Supplementary movie S5 (AVI 797 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Yolamanova, M., Meier, C., Shaytan, A. et al. Peptide nanofibrils boost retroviral gene transfer and provide a rapid means for concentrating viruses. Nature Nanotech 8, 130–136 (2013). https://doi.org/10.1038/nnano.2012.248

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nnano.2012.248

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