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.

  • Protocol
  • Published:

Optimized retroviral transduction of mouse T cells for in vivo assessment of gene function

Abstract

Retroviral (RV) expression of genes of interest (GOIs) is an invaluable tool and has formed the foundation of cellular engineering for adoptive cell therapy in cancer and other diseases. However, monitoring of transduced T cells long term (weeks to months) in vivo remains challenging because of the low frequency and often poor durability of transduced T cells over time when transferred without enrichment. Traditional methods often require additional overnight in vitro culture after transduction. Moreover, in vitro-generated effector CD8+ T cells enriched by sorting often have reduced viability, making it difficult to monitor the fate of transferred cells in vivo. Here, we describe an optimized mouse CD8+ T-cell RV transduction protocol that uses simple and rapid Percoll density centrifugation to enrich RV-susceptible activated CD8+ T cells. Percoll density centrifugation is simple, can be done on the day of transduction, requires minimal time, has low reagent costs and improves cell recovery (up to 60%), as well as the frequency of RV-transduced cells (sixfold over several weeks in vivo as compared with traditional methods). We have used this protocol to assess the long-term stability of CD8+ T cells after RV transduction by comparing the durability of T cells transduced with retroviruses expressing each of six commonly used RV reporter genes. Thus, we provide an optimized enrichment and transduction approach that allows long-term in vivo assessment of RV-transduced T cells. The overall procedure from T-cell isolation to RV transduction takes 2 d, and enrichment of activated T cells can be done in 1 h.

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: Schematic overview of retroviral transduction experiments in mouse T cells.
Figure 2: Enrichment of RV-transduced CD8+ T cells before transfer improves the efficiency of retroviral experiments in vivo.
Figure 3: Reduced cell viability after flow-cytometric sorting leads to instability of transferred T cells in vivo.
Figure 4: Optimal number of transferred P14 cells in Percoll density enrichment in LCMV Arm infection.
Figure 5: Optimal timing and facilitator for RV transduction to CD8+ T cells.
Figure 6: Comparing the utility of six different RV reporter genes for the study of CD8+ T-cell stability and memory differentiation in vivo.
Figure 7: Pre-fixation of RV-transduced T cells prevents the loss of RV-derived marker detection.

Similar content being viewed by others

References

  1. Kaech, S.M. & Cui, W. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat. Rev. Immunol. 12, 749–761 (2012).

    Article  CAS  Google Scholar 

  2. Masopust, D. & Schenkel, J.M. The integration of T cell migration, differentiation and function. Nat. Rev. Immunol. 13, 309–320 (2013).

    Article  CAS  Google Scholar 

  3. Wherry, E.J. & Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15, 486–499 (2015).

    Article  CAS  Google Scholar 

  4. Cepko, C. & Pear, W. Overview of the retrovirus transduction system. Curr. Protoc. Mol. Biol. Chapter 9, Unit 9 9 (2001).

  5. Lee, J., Sadelain, M. & Brentjens, R. in Genetic Modification of Hematopoietic Stem Cells: Methods and Protocols (ed. Baum, C.) 83–96 (Humana Press, 2009).

  6. Zhong, S., Malecek, K., Perez-Garcia, A. & Krogsgaard, M. Retroviral transduction of T-cell receptors in mouse T-cells. J. Vis. Exp. 2010, 2307 (2010).

    Google Scholar 

  7. Kerkar, S.P. et al. Genetic engineering of murine CD8+ and CD4+ T cells for preclinical adoptive immunotherapy studies. J. Immunother. 34, 343–352 (2011).

    Article  CAS  Google Scholar 

  8. Kao, C. et al. Transcription factor T-bet represses expression of the inhibitory receptor PD-1 and sustains virus-specific CD8(+) T cell responses during chronic infection. Nat. Immunol. 12, 663–671 (2011).

    Article  CAS  Google Scholar 

  9. Kurachi, M. et al. The transcription factor BATF operates as an essential differentiation checkpoint in early effector CD8(+) T cells. Nat. Immunol. 15, 373–383 (2014).

    Article  CAS  Google Scholar 

  10. Bengsch, B. et al. Bioenergetic insufficiencies due to metabolic alterations regulated by the inhibitory receptor PD-1 are an early driver of CD8(+) T cell exhaustion. Immunity 45, 358–373 (2016).

    Article  CAS  Google Scholar 

  11. Lamers, C.H., Willemsen, R.A., Luider, B.A., Debets, R. & Bolhuis, R.L. Protocol for gene transduction and expansion of human T lymphocytes for clinical immunogene therapy of cancer. Cancer Gene Ther. 9, 613–623 (2002).

    Article  CAS  Google Scholar 

  12. Parente-Pereira, A.C., Wilkie, S., van der Stegen, S.J.C., Davies, D.M. & Maher, J. Use of retroviral-mediated gene transfer to deliver and test function of chimeric antigen receptors in human T-cells. J. Biol. Methods 1, e7 (2014).

    Article  Google Scholar 

  13. Cepko, C. & Pear, W. Retrovirus infection of cells in vitro and in vivo. Curr. Protoc. Mol. Biol. Chapter 9, Unit 9.14 (2001).

  14. Engels, B. et al. Retroviral vectors for high-level transgene expression in T lymphocytes. Hum. Gene Ther. 14, 1155–1168 (2003).

    Article  CAS  Google Scholar 

  15. Zhang, T., Tsang, T.C. & Harris, D.T. Efficient transduction of murine primary T cells requires a combination of high viral titer, preferred tropism, and proper timing of transduction. J. Hematother. Stem Cell Res. 12, 123–130 (2003).

    Article  CAS  Google Scholar 

  16. Odorizzi, P.M., Pauken, K.E., Paley, M.A., Sharpe, A. & Wherry, E.J. Genetic absence of PD-1 promotes accumulation of terminally differentiated exhausted CD8+ T cells. J. Exp. Med. 212, 1125–1137 (2015).

    Article  CAS  Google Scholar 

  17. Marzo, A.L. et al. Initial T cell frequency dictates memory CD8+ T cell lineage commitment. Nat. Immunol. 6, 793–799 (2005).

    Article  CAS  Google Scholar 

  18. Ansari, A.M. et al. Cellular GFP toxicity and immunogenicity: potential confounders in in vivo cell tracking experiments. Stem Cell Rev. 12, 553–559 (2016).

    Article  CAS  Google Scholar 

  19. D′Souza., W.N. & Hedrick, S.M. Cutting edge: latecomer CD8 T cells are imprinted with a unique differentiation program. J. Immun. 177, 777–781 (2006).

    Article  Google Scholar 

  20. Mozo, L., Rivas, D., Zamorano, J. & Gutierrez, C. Differential expression of IL-4 receptors in human T and B lymphocytes. J. Immunol. 150, 4261–4269 (1993).

    CAS  PubMed  Google Scholar 

  21. Pistoia, V. et al. Production of hematopoietic growth factors by human B lymphocytes: mechanisms and possible implications. Stem Cells 11, 150–155 (1993).

    Article  CAS  Google Scholar 

  22. Pertoft, H. Fractionation of cells and subcellular particles with Percoll. J. Biochem. Biophys. Methods 44, 1–30 (2000).

    Article  CAS  Google Scholar 

  23. Holst, J. et al. Generation of T-cell receptor retrogenic mice. Nat. Protoc. 1, 406–417 (2006).

    Article  CAS  Google Scholar 

  24. Haviernik, P., Zhang, Y. & Bunting, K.D. Retroviral transduction of murine hematopoietic stem cells. Methods Mol. Biol. 430, 229–241 (2008).

    Article  CAS  Google Scholar 

  25. Pear, W. Transient transfection methods for preparation of high-titer retroviral supernatants. Curr. Protoc. Mol. Biol. Chapter 9, Unit 9.11 (2001).

  26. Pircher, H., Burki, K., Lang, R., Hengartner, H. & Zinkernagel, R.M. Tolerance induction in double specific T-cell receptor transgenic mice varies with antigen. Nature 342, 559–561 (1989).

    Article  CAS  Google Scholar 

  27. Hagani, A.B., Riviere, I., Tan, C., Krause, A. & Sadelain, M. Activation conditions determine susceptibility of murine primary T-lymphocytes to retroviral infection. J. Gene Med. 1, 341–351 (1999).

    Article  CAS  Google Scholar 

  28. Dupl′akova, N., Dobrev, P.I., Renak, D. & Honys, D. Rapid separation of Arabidopsis male gametophyte developmental stages using a Percoll gradient. Nat. Protoc. 11, 1817–1832 (2016).

    Article  Google Scholar 

  29. Davis, H.E., Rosinski, M., Morgan, J.R. & Yarmush, M.L. Charged polymers modulate retrovirus transduction via membrane charge neutralization and virus aggregation. Biophys. J. 86, 1234–1242 (2004).

    Article  CAS  Google Scholar 

  30. Weber, B.N. et al. A critical role for TCF-1 in T-lineage specification and differentiation. Nature 476, 63–68 (2011).

    Article  CAS  Google Scholar 

  31. Karasawa, S., Araki, T., Nagai, T., Mizuno, H. & Miyawaki, A. Cyan-emitting and orange-emitting fluorescent proteins as a donor/acceptor pair for fluorescence resonance energy transfer. Biochem. J. 381, 307–312 (2004).

    Article  CAS  Google Scholar 

  32. Wu, Y. et al. FOXP3 controls regulatory T cell function through cooperation with NFAT. Cell 126, 375–387 (2006).

    Article  CAS  Google Scholar 

  33. Izon, D.J. et al. Notch1 regulates maturation of CD4+ and CD8+ thymocytes by modulating TCR signal strength. Immunity 14, 253–264 (2001).

    Article  CAS  Google Scholar 

  34. Heinen, A.P. et al. Improved method to retain cytosolic reporter protein fluorescence while staining for nuclear proteins. Cytometry A 85, 621–627 (2014).

    Article  Google Scholar 

  35. Ahmed, R. et al. Selection of genetic variants of lymphocytic choriomeningitis virus in spleens of persistently infected mice. Role in suppression of cytotoxic T lymphocyte response and viral persistence. J. Exp. Med. 160, 521–540 (1984).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M.A. Ali for animal care; S. Adamski for technical assistance; K. Lewy for plasmid work; J.E. Wu for protocol verification; T. Yamada and K. Rome for comments on RV transduction of CD4+ T cells; and W.S. Pear (University of Pennsylvania), A. Bhandoola (Center for Cancer Research, National Cancer Institute), S.L. Reiner (Columbia University) and D.P. Beiting (University of Pennsylvania) for plasmids, cell line, template protocols and comments. This work was supported by German Research Foundation fellowship BE5496/1-1 (to B.B.), National Institutes of Health (NIH) grants F30AI129263 (to O.K.) and AI105343, AI112521, AI115712, AI108545, AI117950, AI117718, AI082630, AI095608 (to E.J.W.), U.S. Broad Agency Announcements Grant HHSN272201100018C (to E.J.W.) and funding from the Parker Institute for Cancer Immunotherapy (to E.J.W.).

Author information

Authors and Affiliations

Authors

Contributions

M.K. and E.J.W. designed the protocol. M.K. and J.K. made the RV stock. J.K. prepared mouse T cells and analyzed flow cytometry data. M.K. optimized density centrifugation. Z.C., J.J. and O.K. optimized in vitro T-cell stimulation and RV transduction methods. M.K., J.K., Z.C., J.J., O.K., B.B., E.S., J.A. and L.M.M. performed the in vivo experiments. J.A. and L.M.M. developed and verified a new fixation method for RV-transduced cells. M.T. and S.U. contributed to the development of a new Kusabira Orange 2 retrovirus. J.K. prepared all figures. M.K. and E.J.W. wrote the manuscript.

Corresponding author

Correspondence to E John Wherry.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Additional two examples of activated CD8+ T cell enrichment with Percoll centrifugation. (Related to Figure 2b,c)

As shown in Fig.1 and 2b,c, in vitro activated CD8+ T cells were enriched at day 1 using 30% and 60% Percoll centrifugation (a and b: repeat 1 at 18 hrs after stimulation, c and d: repeat 2 at 25hrs after stimulation). (a and c) Flow plots that are gated on the 7AAD negative CD8 positive population show enrichment step from input (left) to shortly after Percoll separation at day 1 (right). (b and d) Graph showing recovery of small (‘resting’) and large (‘blast’) size cells in each interface and bottom after Percoll centrifugation. Note that the recovery of large-sized cells (black bar) was 40% (18 hrs-stimulated; Supplementary Fig. 1b), 60% (22 hrs; Fig. 2c), and 43.4% (25 hrs; Supplementary Fig. 1d).

Supplementary Figure 2 Staining and gating strategy to detect LCMV gp33-specific CD8+ T cells in vivo used in this study.

At the indicated time points in each Figure legend, peripheral blood or spleens were harvested and single cell suspensions were prepared using ACK lysis buffer, followed by staining with anti-CD8, CD44, CD45.1, CD45.2 antibodies, and Dbgp33 tetramer. In some experiments, to detect only transferred P14 cells, TCR Vα2 was used instead of Dbgp33 tetramer. Antigen-specific CD8+ T cells are identified by first gating on singlet cells (FS-A and FS-H), and then on live cells (7AAD negative), and lymphocytes (FS-A and SS-A). Antigen–specific CD8+ T cells were identified by successively gating on CD8, Dbgp33 tetramer (or TCR Vα2) and CD44 positive populations. Transferred P14 cells (and endogenous gp33-specific CD8+ T cells in case of Dbgp33 tetramer staining) were further subgated on the basis of CD45.1 and CD45.2 congenic marker expression. RV-reporter expression was analyzed on the final P14 cell gate. In selected experiments, differentiation and function of effector and memory P14 cells were evaluated by additional surface marker staining such as CD127 and KLRG1 and standard intracellular cytokine staining after 5 hrs restimulation with cognate peptide. Representative blood sample used in Figure 2d as no Percoll condition is shown.

Supplementary Figure 3 Small-sized cell frequency at adoptive transfer inversely correlates with RV-transduction efficiency in vivo. (Related to Figure 2a–g)

(a) Experimental design. Donor P14 cells and recipient mice were stimulated in vitro and infected with LCMV Arm at day 0, respectively. At day 1, activated cells were harvested from the flask and analyzed immediately by flow cytometry to determine the ratio of small-sized cells to large-sized cells in ‘Input’ population. An aliquot of activated cells was set aside for a no Percoll control prior to Percoll centrifugation. After Percoll, interface (blasting cells) and bottom (resting cells) cells were separately collected, and a re-mixture of the interface and bottom cells was made. Then the equal numbers of the 4 populations (3x106 cells per well) were transduced with empty-GFP RV. After 4 hrs, aliquots of the 4 populations were checked by flow cytometry (‘transfer’ sample), and 1x105 cells of each population were adoptively transferred to infected mice. RV-transduction efficiency and engraftment of RV-transduced cells were analyzed at day 2 and 8, respectively. (b) Flow plots gated on live P14 cells show the composition of blasting and resting cells in input (left) and 4 populations at transfer (right-left) at day 1, and RV transduction efficiency at day 2 (right-middle), and frequency of RV-transduced cells at day 8 (right-right). (c) Graph showing ‘Retention rate’ of RV-transduced cells among total P14 cells from day 2 (in vitro) to day 8 (blood). Bars show mean ± s.e.m. *p<0.005 (two-tailed t test); NS p>0.05. All data in this figure are from one experiment (n=5 per group). All animal experiments used in this Figure were in accordance with the Institute Animal Care and Use Guidelines for the University of Pennsylvania.

Supplementary Figure 4 Activated CD4+ T cells can be enriched by Percoll density centrifugation. (Related to Figure 2a–c and 5a)

Polyclonal CD4+ T cells were prepared from C57/Bl6 spleens using EasySep Mouse CD4+ T Cell Isolation Kit (STEMCELLS) and stimulated in vitro with anti-CD3ɛ (final 1 μg/mL), anti-CD28 (final 0.5 μg/mL) antibodies and rIL-2 (final 100 U/mL) as shown in Fig.1. CD4+ T cells were spin-transduced with empty-GFP RV using Polybrene (final 4 μg/mL) and tissue culture treated plates at day 1 (a) or indicated timing (d) after stimulation without enrichment of blasting cells, and analyzed for GFP expression one day after RV-transduction. Some of CD4+ T cells were processed by density centrifugation with 30% and 60% Percoll layers at day 1 (b, c). Data are representative of two independent experiments (one technical replicate per experiment). (a) Small sized ‘resting’ CD4+ T cells are mostly RV negative. (b) Density centrifugation enriches activated ‘blasting’ CD4+ T cells at day 1. Flow plots that are gated on 7-AAD negative CD4 positive population show enrichment step from input (left) to shortly after Percoll separation at day 1 (right). (c) Graph showing recovery of small (‘resting’) and large (‘blast’) size CD4+ cells in each interface and bottom after Percoll centrifugation. (d) RV transduction efficiency to CD4+ cells peaks at 48 hours after in vitro stimulation.

Supplementary information

Supplementary Text and Figures

Supplementary Table 1 and Supplementary Figures 1–4. (PDF 996 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kurachi, M., Kurachi, J., Chen, Z. et al. Optimized retroviral transduction of mouse T cells for in vivo assessment of gene function. Nat Protoc 12, 1980–1998 (2017). https://doi.org/10.1038/nprot.2017.083

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2017.083

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