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:

Continuous human iPSC-macrophage mass production by suspension culture in stirred tank bioreactors

Abstract

Macrophages derived from human induced pluripotent stem cells (iPSCs) have the potential to enable the development of cell-based therapies for numerous disease conditions. We here provide a detailed protocol for the mass production of iPSC-derived macrophages (iPSC-Mac) in scalable suspension culture on an orbital shaker or in stirred-tank bioreactors (STBRs). This strategy is straightforward, robust and characterized by the differentiation of primed iPSC aggregates into ‘myeloid-cell-forming-complex’ intermediates by means of a minimal cytokine cocktail. In contrast to the ‘batch-like differentiation approaches’ established for other iPSC-derived lineages, myeloid-cell-forming-complex-intermediates are stably maintained in suspension culture and continuously generate functional and highly pure iPSC-Mac. Employing a culture volume of 120 ml in the STBR platform, ~1–4 × 107 iPSC-Mac can be harvested at weekly intervals for several months. The STBR technology allows for real-time monitoring of crucial process parameters such as biomass, pH, dissolved oxygen, and nutrition levels; the system also promotes systematic process development, optimization and linear upscaling. The process duration, from the expansion of iPSC until the first iPSC-Mac harvest, is 28 d. Successful application of the protocol requires expertise in pluripotent stem cell culture, differentiation and analytical methods, such as flow cytometry. Fundamental know-how in biotechnology is also advantageous to run the process in the STBR platform. The continuous, scalable production of well-defined iPSC-Mac populations is highly relevant to various fields, ranging from developmental biology, immunology and cell therapies to industrial applications for drug safety and discovery.

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 representation of the differentiation platform.
Fig. 2: Morphology of human iPSCs in 2D culture.
Fig. 3: Morphology of primed aggregates.
Fig. 4: Photography of the assembled bioreactor (related to Box 1).
Fig. 5: Production of iPSC-Mac by MCFCs.
Fig. 6: Harvest efficiency and process monitoring of the STBR production platform.
Fig. 7: Analysis of iPSC-Mac by flow cytometry (related to Box 4).
Fig. 8: Phagocytosis of bioparticles by iPSC-Mac (related to Box 5).

Similar content being viewed by others

Data availability statement

The raw data for Figs. 7 and 8 are available under FlowRepository IDs FR-FCM-Z4WC (Fig. 7) and FR-FCM-Z4WE (Fig. 8)). Source data are provided with this paper.

References

  1. Kempf, H. & Zweigerdt, R. Scalable cardiac differentiation of pluripotent stem cells using specific growth factors and small molecules. Adv. Biochem. Eng. Biotechnol. 163, 39–69 (2018).

    CAS  PubMed  Google Scholar 

  2. Knorr, D. A. et al. Clinical-scale derivation of natural killer cells from human pluripotent stem cells for cancer therapy. Stem Cells Transl. Med. 2, 274–283 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Ito, Y. et al. Turbulence activates platelet biogenesis to enable clinical scale ex vivo production. Cell 174, 636–648.e618 (2018).

    CAS  PubMed  Google Scholar 

  4. Wynn, T. A., Chawla, A. & Pollard, J. W. Macrophage biology in development, homeostasis and disease. Nature 496, 445–455 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Hetzel, M., Ackermann, M. & Lachmann, N. Beyond “big eaters”: the versatile role of alveolar macrophages in health and disease. Int. J. Mol. Sci. https://doi.org/10.3390/ijms22073308 (2021).

  6. Singanayagam, A. & Triantafyllou, E. Macrophages in chronic liver failure: diversity, plasticity and therapeutic targeting. Front. Immunol. 12, 661182 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Ginhoux, F. & Guilliams, M. Tissue-resident macrophage ontogeny and homeostasis. Immunity 44, 439–449 (2016).

    CAS  PubMed  Google Scholar 

  8. Haake, K., Ackermann, M. & Lachmann, N. Concise review: towards the clinical translation of induced pluripotent stem cell-derived blood cells-ready for take-off. Stem Cells Transl. Med. 8, 332–339 (2019).

    PubMed  Google Scholar 

  9. Lee, C. Z. W., Kozaki, T. & Ginhoux, F. Studying tissue macrophages in vitro: are iPSC-derived cells the answer? Nat. Rev. Immunol. 18, 716–725 (2018).

    CAS  PubMed  Google Scholar 

  10. Ackermann, M., Dragon, A. C. & Lachmann, N. The immune-modulatory properties of iPSC-derived antigen-presenting cells. Transfus. Med. Hemother. 47, 444–453 (2020).

    PubMed  PubMed Central  Google Scholar 

  11. Happle, C. et al. Pulmonary transplantation of human induced pluripotent stem cell-derived macrophages ameliorates pulmonary alveolar proteinosis. Am. J. Respir. Crit. Care Med. 198, 350–360 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Xu, R. et al. Human iPSC-derived mature microglia retain their identity and functionally integrate in the chimeric mouse brain. Nat. Commun. 11, 1577 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Ackermann, M. et al. Bioreactor-based mass production of human iPSC-derived macrophages enables immunotherapies against bacterial airway infections. Nat. Commun. 9, 5088 (2018).

    PubMed  PubMed Central  Google Scholar 

  14. Karlsson, K. R. et al. Homogeneous monocytes and macrophages from human embryonic stem cells following coculture-free differentiation in M-CSF and IL-3. Exp. Hematol. 36, 1167–1175 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. van Wilgenburg, B., Browne, C., Vowles, J. & Cowley, S. A. Efficient, long term production of monocyte-derived macrophages from human pluripotent stem cells under partly-defined and fully-defined conditions. PLoS One 8, e71098 (2013).

    PubMed  PubMed Central  Google Scholar 

  16. Lachmann, N. et al. Large-scale hematopoietic differentiation of human induced pluripotent stem cells provides granulocytes or macrophages for cell replacement therapies. Stem Cell Rep. 4, 282–296 (2015).

    CAS  Google Scholar 

  17. Ackermann, M. et al. A 3D iPSC-differentiation model identifies interleukin-3 as a regulator of early human hematopoietic specification. Haematologica 106, 1354–1367 (2021).

    CAS  PubMed  Google Scholar 

  18. Dreyer, A. K. et al. TALEN-mediated functional correction of X-linked chronic granulomatous disease in patient-derived induced pluripotent stem cells. Biomaterials 69, 191–200 (2015).

    CAS  PubMed  Google Scholar 

  19. Lachmann, N. et al. Gene correction of human induced pluripotent stem cells repairs the cellular phenotype in pulmonary alveolar proteinosis. Am. J. Respir. Crit. Care Med. 189, 167–182 (2014).

    CAS  PubMed  Google Scholar 

  20. Haake, K. et al. Patient iPSC-derived macrophages to study inborn errors of the IFN-γ responsive pathway. Cells https://doi.org/10.3390/cells9020483 (2020).

  21. Neehus, A. L. et al. Impaired IFNγ-signaling and mycobacterial clearance in IFNγR1-deficient human iPSC-derived macrophages. Stem Cell Rep. 10, 7–16 (2018).

    CAS  Google Scholar 

  22. Dannenmann, B. et al. iPSC modeling of stage-specific leukemogenesis reveals BAALC as a key oncogene in severe congenital neutropenia. Cell Stem Cell https://doi.org/10.1016/j.stem.2021.03.023 (2021).

  23. Dannenmann, B. et al. Human iPSC-based model of severe congenital neutropenia reveals elevated UPR and DNA damage in CD34+ cells preceding leukemic transformation. Exp. Hematol. 71, 51–60 (2019).

    CAS  PubMed  Google Scholar 

  24. Le Voyer, T. et al. Inherited deficiency of stress granule ZNFX1 in patients with monocytosis and mycobacterial disease. Proc. Natl Acad. Sci. USA https://doi.org/10.1073/pnas.2102804118 (2021).

  25. Makaryan, V. et al. Elastase inhibitors as potential therapies for ELANE-associated neutropenia. J. Leukoc. Biol. 102, 1143–1151 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Pittermann, E. et al. Gene correction of HAX1 reversed Kostmann disease phenotype in patient-specific induced pluripotent stem cells. Blood Adv. 1, 903–914 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Kropp, C. et al. Impact of feeding strategies on the scalable expansion of human pluripotent stem cells in single-use stirred tank bioreactors. Stem Cells Transl. Med. 5, 1289–1301 (2016).

    PubMed  PubMed Central  Google Scholar 

  28. Manstein, F. et al. High density bioprocessing of human pluripotent stem cells by metabolic control and in silico modeling. Stem Cells Transl. Med. https://doi.org/10.1002/sctm.20-0453 (2021).

  29. Halloin, C. et al. Continuous WNT control enables advanced hPSC cardiac processing and prognostic surface marker identification in chemically defined suspension culture. Stem Cell Rep. https://doi.org/10.1016/j.stemcr.2019.09.001 (2019).

  30. Kempf, H., Kropp, C., Olmer, R., Martin, U. & Zweigerdt, R. Cardiac differentiation of human pluripotent stem cells in scalable suspension culture. Nat. Protoc. 10, 1345–1361 (2015).

    CAS  PubMed  Google Scholar 

  31. Olmer, R. et al. Differentiation of human pluripotent stem cells into functional endothelial cells in scalable suspension culture. Stem Cell Rep. 10, 1657–1672 (2018).

    CAS  Google Scholar 

  32. Sahabian, A., Dahlmann, J., Martin, U. & Olmer, R. Production and cryopreservation of definitive endoderm from human pluripotent stem cells under defined and scalable culture conditions. Nat. Protoc. 16, 1581–1599 (2021).

    CAS  PubMed  Google Scholar 

  33. Buchrieser, J., James, W. & Moore, M. D. Human induced pluripotent stem cell-derived macrophages share ontogeny with MYB-independent tissue-resident macrophages. Stem Cell Rep. 8, 334–345 (2017).

    CAS  Google Scholar 

  34. Rafiei Hashtchin, A. et al. Human iPSC-derived macrophages for efficient Staphylococcus aureus clearance in a murine pulmonary infection model. Blood Adv. https://doi.org/10.1182/bloodadvances.2021004853 (2021).

  35. Fattorelli, N. et al. Stem-cell-derived human microglia transplanted into mouse brain to study human disease. Nat. Protoc. 16, 1013–1033 (2021).

    CAS  PubMed  Google Scholar 

  36. Capotondo, A. et al. Intracerebroventricular delivery of hematopoietic progenitors results in rapid and robust engraftment of microglia-like cells. Sci. Adv. 3, e1701211 (2017).

    PubMed  PubMed Central  Google Scholar 

  37. Bird, T. G. et al. Bone marrow injection stimulates hepatic ductular reactions in the absence of injury via macrophage-mediated TWEAK signaling. Proc. Natl Acad. Sci. USA 110, 6542–6547 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Moroni, F. et al. Safety profile of autologous macrophage therapy for liver cirrhosis. Nat. Med. 25, 1560–1565 (2019).

    CAS  PubMed  Google Scholar 

  39. Klichinsky, M. et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 38, 947–953 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Ackermann, M. et al. Restored macrophage function ameliorates disease pathophysiology in a mouse model for IL10 receptor deficient very early onset inflammatory bowel disease. J. Crohns Colitis https://doi.org/10.1093/ecco-jcc/jjab031 (2021).

  41. Ackermann, M. et al. Ex vivo generation of genetically modified macrophages from human induced pluripotent stem cells. Transfus. Med. Hemother. 44, 135–142 (2017).

    PubMed  PubMed Central  Google Scholar 

  42. Lopez-Yrigoyen, M. et al. Genetic programming of macrophages generates an in vitro model for the human erythroid island niche. Nat. Commun. 10, 881 (2019).

    PubMed  PubMed Central  Google Scholar 

  43. Ackermann, M. et al. A 3D iPSC-differentiation model identifies interleukin-3 as a regulator of early human hematopoietic specification. Haematologica https://doi.org/10.3324/haematol.2019.228064 (2020).

  44. Bernecker, C. et al. Enhanced ex vivo generation of erythroid cells from human induced pluripotent stem cells in a simplified cell culture system with low cytokine support. Stem Cells Dev. 28, 1540–1551 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Choi, K. D., Vodyanik, M. & Slukvin, I. I. Hematopoietic differentiation and production of mature myeloid cells from human pluripotent stem cells. Nat. Protoc. 6, 296–313 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Doulatov, S. et al. Induction of multipotential hematopoietic progenitors from human pluripotent stem cells via respecification of lineage-restricted precursors. Cell Stem Cell 13, 459–470 (2013).

    CAS  PubMed  Google Scholar 

  47. Vo, L. T. et al. Regulation of embryonic haematopoietic multipotency by EZH1. Nature 553, 506–510 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Gutbier, S. et al. Large-scale production of human iPSC-derived macrophages for drug screening. Int. J. Mol. Sci. https://doi.org/10.3390/ijms21134808 (2020).

  49. Lancaster, M. A. & Knoblich, J. A. Organogenesis in a dish: modeling development and disease using organoid technologies. Science 345, 1247125 (2014).

    PubMed  Google Scholar 

  50. Cao, X. et al. Differentiation and functional comparison of monocytes and macrophages from hiPSCs with peripheral blood derivatives. Stem Cell Rep. 12, 1282–1297 (2019).

    CAS  Google Scholar 

  51. Ackermann, M. et al. Promoter and lineage independent anti-silencing activity of the A2 ubiquitous chromatin opening element for optimized human pluripotent stem cell-based gene therapy. Biomaterials 35, 1531–1542 (2014).

    CAS  PubMed  Google Scholar 

  52. Hong, D. et al. Human-induced pluripotent stem cell-derived macrophages and their immunological function in response to tuberculosis infection. Stem Cell Res. Ther. 9, 49 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Lopez-Yrigoyen, M. et al. A human iPSC line capable of differentiating into functional macrophages expressing ZsGreen: a tool for the study and in vivo tracking of therapeutic cells. Philos. Trans. R. Soc. Lond. B Biol. Sci. https://doi.org/10.1098/rstb.2017.0219 (2018).

  54. Neehus, A. L. et al. Impaired respiratory burst contributes to infections in PKCδ-deficient patients. J. Exp. Med https://doi.org/10.1084/jem.20210501 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Kuo, H. H. et al. Negligible-cost and weekend-free chemically defined human ipsc culture. Stem Cell Rep. 14, 256–270 (2020).

    CAS  Google Scholar 

  56. Ishida, T. et al. Live-cell imaging of macrophage phagocytosis of asbestos fibers under fluorescence microscopy. Genes Environ. 41, 14 (2019).

    PubMed  PubMed Central  Google Scholar 

  57. Lewis, L. E., Bain, J. M., Okai, B., Gow, N. A. & Erwig, L. P. Live-cell video microscopy of fungal pathogen phagocytosis. J. Vis. Exp. https://doi.org/10.3791/50196 (2013).

Download references

Acknowledgements

The authors thank D. Kloos, T. Buchegger and L. Bach for technical support. The work received funding from the German Research Foundation (Deutsche Forschungsgemeinschaft, DFG): Cluster of Excellence REBIRTH EXC 62/3 (R.Z. and N.L.), ZW64/4-1, ZW 64/4-2, KFO311/ZW64/7-1 (R.Z.); the German Ministry for Education and Science (Bundesministerium für Bildung und Forschung, BMBF): 01EK1602A (R.Z. and N.L.) 13N14086, 01EK1601A, 655 13XP5092B, 031L0249 (R.Z.), ‘Förderung aus Mitteln des Niedersächsischen Vorab’ (grant ZN3340) (R.Z. and N.L.), the Else Kröner-Fresenius-Stiftung (EKFS; 2016_A146) (M.A.). The work also received funding from Hannover Medical School Transplantation Center (Tx Center) (N.L.) and the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) (N.L.). This project has received funding from the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation program (grant agreement no. 852178).

Author information

Authors and Affiliations

Authors

Contributions

M.A., H.K., R.Z. and N.L. developed the protocol; M.A., A.R.H. and F.M. performed the experiments and analyzed the data; M.A., A.R.H., F.M., M.C.O., R.Z. and N.L wrote the manuscript; all authors approved the final paper.

Corresponding author

Correspondence to Nico Lachmann.

Ethics declarations

Competing interests

This work is included in a patent application. M.A., H.K., R.Z. and N.L. are authors of the patent application (European patent application number PCT/EP2018/061574) entitled ‘Stem-cell derived myeloid cells, generation and use thereof’. The priority date of the application is 4 May 2017. H.K. is an employee of Novo Nordisk A/S, Måløv, Denmark, and N.L. and R.Z. receive funding from Novo Nordisk A/S, Måløv, Denmark.

Peer review

Peer review information

Nature Protocols thanks Dan Kaufman, Lesley Forrester and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Ackermann, M. et al. Nat. Commun. 9, 5088 (2018): https://doi.org/10.1038/s41467-018-07570-7

Ackermann, M et al. Haematologica 106, 1354–1367 (2021): https://doi.org/10.3324/haematol.2019.228064

Rafiei Hashtchin, A. et al. Blood Adv. 5, 5190–5201 (2021): https://doi.org/10.1182/bloodadvances.2021004853

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ackermann, M., Rafiei Hashtchin, A., Manstein, F. et al. Continuous human iPSC-macrophage mass production by suspension culture in stirred tank bioreactors. Nat Protoc 17, 513–539 (2022). https://doi.org/10.1038/s41596-021-00654-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-021-00654-7

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: 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