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:

Genetic manipulation and immortalized culture of ex vivo primary human germinal center B cells

Abstract

Next-generation sequencing has transformed our knowledge of the genetics of lymphoid malignancies. However, limited experimental systems are available to model the functional effects of these genetic changes and their implications for therapy. The majority of mature B-cell malignancies arise from the germinal center (GC) stage of B-cell differentiation. Here we describe a detailed protocol for the purification and ex vivo expansion of primary, nonmalignant human GC B cells. We present methodology for the high-efficiency transduction of these cells to enable combinatorial expression of putative oncogenes. We also describe alternative approaches for CRISPR–Cas9-mediated deletion of putative tumor suppressors. Mimicking genetic changes commonly found in lymphoid malignancies leads to immortalized growth in vitro, while engraftment into immunodeficient mice generates genetically customized, synthetic models of human lymphoma. The protocol is simple and inexpensive and can be implemented in any laboratory with access to standard cell culture and animal facilities. It can be easily scaled up to enable high-throughput screening and thus provides a versatile platform for the functional interrogation of lymphoma genomic data.

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

Fig. 1: Schematic overview of the expansion, immortalization and use of human GC B cells cultured on FDC-like feeder cells.
Fig. 2: Summary of the GC B-cell purification strategy.
Fig. 3: Example of successful GC B-cell enrichment.
Fig. 4: Examples of the use of YK6-CD40Lg-IL21 and modified feeder systems.
Fig. 5: Gene knockout using virally transduced Cas9 and gRNA.
Fig. 6: Gene knockout using transient nucleofection of RNP complexes.
Fig. 7: Typical histological and immunohistochemical appearances of an example ‘synthetic’ human tumor.

Similar content being viewed by others

Data availability

All raw data underlying the figures are provided as source data files. Other examples of data can be made available upon reasonable request. Source data are provided with this paper.

References

  1. Basso, K. & Dalla-Favera, R. Germinal centres and B cell lymphomagenesis. Nat. Rev. Immunol. 15, 172–184 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Basso, K. & Dalla-Favera, R. BCL6: master regulator of the germinal center reaction and key oncogene in B cell lymphomagenesis. Adv. Immunol. 105, 193–210 (2010).

    Article  CAS  PubMed  Google Scholar 

  3. Shaffer, A. L. 3rd, Young, R. M. & Staudt, L. M. Pathogenesis of human B cell lymphomas. Ann. Rev. Immunol. 30, 565–610 (2012).

    Article  CAS  Google Scholar 

  4. Morin, R. D. et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 476, 298–303 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Pasqualucci, L. et al. Analysis of the coding genome of diffuse large B-cell lymphoma. Nat. Genet. 43, 830–837 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lohr, J. G. et al. Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing. Proc. Natl Acad. Sci. USA 109, 3879–3884 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Zhang, J. et al. Genetic heterogeneity of diffuse large B-cell lymphoma. Proc. Natl Acad. Sci, USA 110, 1398–1403 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Reddy, A. et al. Genetic and functional drivers of diffuse large B Cell Lymphoma. Cell 171, 481–494 e415 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Schmitz, R. et al. Genetics and pathogenesis of diffuse large B-cell lymphoma. N. Engl. J. Med. 378, 1396–1407 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chapuy, B. et al. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat. Med. 24, 679–690 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Karube, K. et al. Integrating genomic alterations in diffuse large B-cell lymphoma identifies new relevant pathways and potential therapeutic targets. Leukemia 32, 675–684 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Arthur, S. E. et al. Genome-wide discovery of somatic regulatory variants in diffuse large B-cell lymphoma. Nat. Commun. 9, 4001 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Morin, R. D. et al. Mutational and structural analysis of diffuse large B-cell lymphoma using whole-genome sequencing. Blood 122, 1256–1265 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Schmitz, R. et al. Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics. Nature 490, 116–120 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lopez, C. et al. Genomic and transcriptomic changes complement each other in the pathogenesis of sporadic Burkitt lymphoma. Nat. Commun. 10, 1459 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Richter, J. et al. Recurrent mutation of the ID3 gene in Burkitt lymphoma identified by integrated genome, exome and transcriptome sequencing. Nat. Genet. 44, 1316–1320 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Panea, R. I. et al. The whole-genome landscape of Burkitt lymphoma subtypes. Blood 134, 1598–1607 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Okosun, J. et al. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat. Genet. 46, 176–181 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Pasqualucci, L. et al. Genetics of follicular lymphoma transformation. Cell Rep. 6, 130–140 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kridel, R. et al. Histological transformation and progression in follicular lymphoma: a clonal evolution study. PLoS Med. 13, e1002197 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Hahn, W. C. et al. Creation of human tumour cells with defined genetic elements. Nature 400, 464–468 (1999).

    Article  CAS  PubMed  Google Scholar 

  22. Caeser, R. et al. Genetic modification of primary human B cells to model high-grade lymphoma. Nat. Commun. 10, 4543 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Pound, J. D. & Gordon, J. Maintenance of human germinal center B cells in vitro. Blood 89, 919–928 (1997).

    Article  CAS  PubMed  Google Scholar 

  24. Arpin, C. et al. Generation of memory B cells and plasma cells in vitro. Science 268, 720–722 (1995).

    Article  CAS  PubMed  Google Scholar 

  25. Banchereau, J. et al. The CD40 antigen and its ligand. Ann. Rev. Immunol. 12, 881–922 (1994).

    Article  CAS  Google Scholar 

  26. Kim, H. S., Zhang, X., Klyushnenkova, E. & Choi, Y. S. Stimulation of germinal center B lymphocyte proliferation by an FDC-like cell line, HK. J. Immunol. 155, 1101–1109 (1995).

    CAS  PubMed  Google Scholar 

  27. Ding, B. B., Bi, E., Chen, H., Yu, J. J. & Ye, B. H. IL-21 and CD40L synergistically promote plasma cell differentiation through upregulation of Blimp-1 in human B cells. J. Immunol. 190, 1827–1836 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Linterman, M. A. et al. IL-21 acts directly on B cells to regulate Bcl-6 expression and germinal center responses. J. Exp. Med. 207, 353–363 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zotos, D. et al. IL-21 regulates germinal center B cell differentiation and proliferation through a B cell-intrinsic mechanism. J. Exp. Med. 207, 365–378 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Christodoulopoulos, I. & Cannon, P. M. Sequences in the cytoplasmic tail of the gibbon ape leukemia virus envelope protein that prevent its incorporation into lentivirus vectors. J. Virol. 75, 4129–4138 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mock, U., Thiele, R., Uhde, A., Fehse, B. & Horn, S. Efficient lentiviral transduction and transgene expression in primary human B cells. Hum. Gene Ther. Methods 23, 408–415 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Gong, C. et al. Sequential inverse dysregulation of the RNA helicases DDX3X and DDX3Y facilitates MYC-driven lymphomagenesis. Preprint at SSRN https://doi.org/10.2139/ssrn.3520953 (2020).

  33. Nojima, T. et al. In-vitro derived germinal centre B cells differentially generate memory B or plasma cells in vivo. Nat. Commun. 2, 465 (2011).

    Article  PubMed  Google Scholar 

  34. Sommermann, T. et al. Functional interplay of Epstein-Barr virus oncoproteins in a mouse model of B cell lymphomagenesis. Proc. Natl Acad. Sci. USA 117, 14421–14432 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Victora, G. D. et al. Identification of human germinal center light and dark zone cells and their relationship to human B-cell lymphomas. Blood 120, 2240–2248 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kjeldsen, M. K. et al. Multiparametric flow cytometry for identification and fluorescence activated cell sorting of five distinct B-cell subpopulations in normal tonsil tissue. Am. J. Clin. Pathol. 136, 960–969 (2011).

    Article  PubMed  Google Scholar 

  37. Kwakkenbos, M. J. et al. Generation of stable monoclonal antibody-producing B cell receptor-positive human memory B cells by genetic programming. Nat. Med. 16, 123–128 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. Stevens, S. J. et al. Monitoring of epstein-barr virus DNA load in peripheral blood by quantitative competitive PCR. J. Clin. Microbiol. 37, 2852–2857 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Tzelepis, K. et al. A CRISPR dropout screen identifies genetic vulnerabilities and therapeutic targets in acute myeloid leukemia. Cell Rep. 17, 1193–1205 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

D.H. was personally supported by a Clinician Scientist Fellowship from the Medical Research Council (MR/M008584/1). Research in the Hodson laboratory is supported by the Kay Kendall Leukaemia Fund, The Addenbrooke’s Charitable Trust and the Evelyn Trust. The Hodson laboratory receives core funding from Wellcome (203151/Z/16/Z) and MRC to the Wellcome-MRC Cambridge Stem Cell Institute and from CRUK to the CRUK Cambridge Centre (A25117). We thank J. Beswick, A. Mitchel and N. Jonas from the ENT Department at Addenbrooke’s Hospital, Cambridge for their assistance in the collection of primary tonsil tissue. We are grateful to K. Elston and J. Baxter from the Cambridge Blood and Stem Cell Bank for collection and storage of primary tonsils tissue and to the staff of the Central Biomedical Services for animal housing and care. We thank A. Weiss for expert technical advice. This research was supported by the Cambridge NIHR BRC Cell Phenotyping Hub and we wish to thank all members of the flow cytometry core for their advice and support in flow cytometry.

Author information

Authors and Affiliations

Authors

Contributions

R.C., J.G., M.D.R., C.G. and D.H. contributed to the development and optimization of the protocols described in this manuscript. R.C. and D.H. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Daniel J. Hodson.

Ethics declarations

Competing interests

R.C.: consultancy for Karus Therapeutics. D.H.: research funding from Gilead Sciences. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Protocols thanks Wolfgang Hammerschmidt and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Key references using this protocol

Caeser, R. et al. Nat. Commun. 10, 4543 (2019): https://doi.org/10.1038/s41467-019-12494-x

Sommermann, T. et al. Proc. Natl Acad. Sci. USA 117, 14421–14432 (2020): https://doi.org/10.1073/pnas.1921139117

Supplementary information

Source data

Source Data Fig. 4

Raw numbers.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Caeser, R., Gao, J., Di Re, M. et al. Genetic manipulation and immortalized culture of ex vivo primary human germinal center B cells. Nat Protoc 16, 2499–2519 (2021). https://doi.org/10.1038/s41596-021-00506-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-021-00506-4

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: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer