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:

Standardized production of hPSC-derived cardiomyocyte aggregates in stirred spinner flasks

Abstract

A promising cell-therapy approach for heart failure aims at differentiating human pluripotent stem cells (hPSCs) into functional cardiomyocytes (CMs) in vitro to replace the disease-induced loss of patients’ heart muscle cells in vivo. But many challenges remain for the routine clinical application of hPSC-derived CMs (hPSC-CMs), including good manufacturing practice (GMP)-compliant production strategies. This protocol describes the efficient generation of hPSC-CM aggregates in suspension culture, emphasizing process simplicity, robustness and GMP compliance. The strategy promotes clinical translation and other applications that require large numbers of CMs. Using a simple spinner-flask platform, this protocol is applicable to a broad range of users with general experience in handling hPSCs without extensive know-how in biotechnology. hPSCs are expanded in monolayer to generate the required cell numbers for process inoculation in suspension culture, followed by stirring-controlled formation of cell-only aggregates at a 300-ml scale. After 48 h at checkpoint (CP) 0, chemically defined cardiac differentiation is induced by WNT-pathway modulation through use of the glycogen-synthase kinase-3 inhibitor CHIR99021 (WNT agonist), which is replaced 24 h later by the chemical WNT-pathway inhibitor IWP-2. The exact application of the described process parameters is important to ensure process efficiency and robustness. After 10 d of differentiation (CP I), the production of ≥100 × 106 CMs is expected. Moreover, to ‘uncouple’ cell production from downstream applications, continuous maintenance of CM aggregates for up to 35 d in culture (CP II) is demonstrated without a reduction in CM content, supporting downstream logistics while potentially overcoming the requirement for cryopreservation.

Key points

  • We present a protocol for the efficient generation of hPSC-CM aggregates in suspension culture, emphasizing process simplicity, robustness and GMP compliance. The strategy promotes clinical translation and other applications that require large numbers of CMs.

  • This protocol uses a simple spinner-flask platform, making it accessible to users experienced in the handling of hPSCs but without extensive experience in biotechnology. This enables straightforward adaptation by many laboratories without bioprocessing experience.

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 display of key steps and timelines of the protocol and the applied culture strategy and vessels.
Fig. 2: Typical appearance of a sub-confluent hPSC culture (hiPSC line Phoenix) grown for 3 d in monolayer before splitting on respective culture matrices.
Fig. 3: Exemplified results expected for quality assurance at CP 0.
Fig. 4: Establishing a viability assay for whole aggregate analysis.
Fig. 5: Typical outcome of QC assessment at CP I and CP II.
Fig. 6: Flow cytometry–based analysis of cells not expressing cTnT (‘non-cardiomyocytes’) at CP I.

Similar content being viewed by others

Data availability

The flow cytometry data are available in the FlowRepository under accession code FR-FCM-Z6K3. All remaining data generated or analyzed during this study are included in this published article and its supplementary files.

References

  1. Laflamme, M. A. & Murry, C. E. Heart regeneration. Nature 473, 326–335 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Kobold, S. et al. A manually curated database on clinical studies involving cell products derived from human pluripotent stem cells. Stem Cell Rep. 15, 546–555 (2020).

    Article  CAS  Google Scholar 

  3. Ilic, D. & Ogilvie, C. Pluripotent stem cells in clinical setting—new developments and overview of current status. Stem Cells 40, 791–801 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Cyranoski, D. ‘Reprogrammed’ stem cells approved to mend human hearts for the first time. Nature 557, 619–620 (2018).

    Article  CAS  PubMed  Google Scholar 

  5. Mallapaty, S. Revealed: two men in China were first to receive pioneering stem-cell treatment for heart disease. Nature 581, 249–250 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Silver, S. E., Barrs, R. W. & Mei, Y. Transplantation of human pluripotent stem cell-derived cardiomyocytes for cardiac regenerative therapy. Front. Cardiovasc. Med. 8, 707890 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Liu, Y.-W. et al. Human embryonic stem cell–derived cardiomyocytes restore function in infarcted hearts of non-human primates. Nat. Biotechnol. 36, 597–605 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. van den Akker, F. et al. Intramyocardial stem cell injection: go(ne) with the flow. Eur. Heart J. 38, 184–186 (2016).

    Google Scholar 

  9. Hogrebe, N. J., Maxwell, K. G., Augsornworawat, P. & Millman, J. R. Generation of insulin-producing pancreatic β cells from multiple human stem cell lines. Nat. Protoc. 16, 4109–4143 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Preininger, M. K., Singh, M. & Xu, C. Cryopreservation of human pluripotent stem cell-derived cardiomyocytes: strategies, challenges, and future directions. Adv. Exp. Med. Biol. 951, 123–135 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. 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. 13, 366–379 (2019).

    Article  CAS  Google Scholar 

  12. Burridge, P. W. et al. Chemically defined generation of human cardiomyocytes. Nat. Methods 11, 855–860 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lian, X. et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc. Natl Acad. Sci. USA 109, E1848–E1857 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lian, X. et al. Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/β-catenin signaling under fully defined conditions. Nat. Protoc. 8, 162–175 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Fonoudi, H. et al. Large-scale production of cardiomyocytes from human pluripotent stem cells using a highly reproducible small molecule-based differentiation protocol. J. Vis. Exp. 2016, 54276 (2016).

    Google Scholar 

  16. Kahn-Krell, A. et al. Bioreactor suspension culture: differentiation and production of cardiomyocyte spheroids from human induced pluripotent stem cells. Front. Bioeng. Biotechnol. 9, 674260 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  17. 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).

    Article  CAS  PubMed  Google Scholar 

  18. Chen, A., Ting, S., Seow, J., Reuveny, S. & Oh, S. Considerations in designing systems for large scale production of human cardiomyocytes from pluripotent stem cells. Stem Cell Res. Ther. 5, 12 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Kempf, H. et al. Controlling expansion and cardiomyogenic differentiation of human pluripotent stem cells in scalable suspension culture. Stem Cell Rep. 3, 1132–1146 (2014).

    Article  CAS  Google Scholar 

  20. Manstein, F. et al. High density bioprocessing of human pluripotent stem cells by metabolic control and in silico modeling. Stem Cells Transl. Med 10, 1063–1080 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Langenberg, K. et al. Controlled stirred tank bioreactors for large-scale manufacture of human iPSC models for cell therapy. Cytotherapy 22, S43 (2020).

    Article  Google Scholar 

  22. Fischer, B. et al. A complete workflow for the differentiation and the dissociation of hiPSC-derived cardiospheres. Stem Cell Res 32, 65–72 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Correia, C. et al. Combining hypoxia and bioreactor hydrodynamics boosts induced pluripotent stem cell differentiation towards cardiomyocytes. Stem Cell Rev. Rep. 10, 786–801 (2014).

    Article  CAS  PubMed  Google Scholar 

  24. Hamad, S. et al. Generation of human induced pluripotent stem cell-derived cardiomyocytes in 2D monolayer and scalable 3D suspension bioreactor cultures with reduced batch-to-batch variations. Theranostics 9, 7222–7238 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sahabian, A. et al. Chemically-defined, xeno-free, scalable production of hPSC-derived definitive endoderm aggregates with multi-lineage differentiation potential. Cells 8, 1571 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ackermann, M. et al. Continuous human iPSC-macrophage mass production by suspension culture in stirred tank bioreactors. Nat. Protoc. 17, 513–539 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Chen, V. C. et al. Development of a scalable suspension culture for cardiac differentiation from human pluripotent stem cells. Stem Cell Res. 15, 365–375 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Shafa, M., Panchalingam, K. M., Walsh, T., Richardson, T. & Baghbaderani, B. A. Computational fluid dynamics modeling, a novel, and effective approach for developing scalable cell therapy manufacturing processes. Biotechnol. Bioeng. 116, 3228–3241 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Kempf, H. et al. Bulk cell density and Wnt/TGFbeta signalling regulate mesendodermal patterning of human pluripotent stem cells. Nat. Commun. 7, 13602 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zweigerdt, R., Olmer, R., Singh, H., Haverich, A. & Martin, U. Scalable expansion of human pluripotent stem cells in suspension culture. Nat. Protoc. 6, 689–700 (2011).

    Article  CAS  PubMed  Google Scholar 

  32. Olmer, R. et al. Suspension culture of human pluripotent stem cells in controlled, stirred bioreactors. Tissue Eng. Part C. Methods 18, 772–784 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kiesslich, S. & Kamen, A. A. Vero cell upstream bioprocess development for the production of viral vectors and vaccines. Biotechnol. Adv. 44, 107608 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ashok, P., Parikh, A., Du, C. & Tzanakakis, E. S. Xenogeneic-free system for biomanufacturing of cardiomyocyte progeny from human pluripotent stem cells. Front. Bioeng. Biotechnol. 8, 571425 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Manstein, F. et al. Protocol process control and in silico modeling strategies for enabling high density culture of human pluripotent stem cells in stirred tank bioreactors. STAR Protoc. 2, 100988 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Gaspari, E. et al. Paracrine mechanisms in early differentiation of human pluripotent stem cells: insights from a mathematical model. Stem Cell Res. 32, 1–7 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Williams, B. et al. Prediction of human induced pluripotent stem cell cardiac differentiation outcome by multifactorial process modeling. Front. Bioeng. Biotechnol. 8, 851 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Floy, M. E. et al. Advances in manufacturing cardiomyocytes from human pluripotent stem cells. Annu. Rev. Chem. Biomol. Eng. 13, 255–278 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kattman, S. J. et al. Stage-specific optimization of activin/nodal and BMP signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell 8, 228–240 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Lundy, S. D., Zhu, W. Z., Regnier, M. & Laflamme, M. A. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells Dev. 22, 1991–2002 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sartiani, L. et al. Developmental changes in cardiomyocytes differentiated from human embryonic stem cells: a molecular and electrophysiological approach. Stem Cells 25, 1136–1144 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Wickramasinghe, N. M. et al. PPARdelta activation induces metabolic and contractile maturation of human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 29, 559–576.e7 (2022).

    Article  CAS  PubMed  Google Scholar 

  43. Krüger, M. et al. Thyroid hormone regulates developmental titin isoform transitions via the phosphatidylinositol-3-kinase/ AKT pathway. Circ. Res. 102, 439–447 (2008).

    Article  PubMed  Google Scholar 

  44. Yang, X. et al. Tri-iodo-l-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J. Mol. Cell. Cardiol. 72, 296–304 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Rog-Zielinska, E. A. et al. Glucocorticoids promote structural and functional maturation of foetal cardiomyocytes: a role for PGC-1α. Cell Death Differ. 22, 1106–1116 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Parikh, S. S. et al. Thyroid and glucocorticoid hormones promote functional T-tubule development in human-induced pluripotent stem cell–derived cardiomyocytes. Circ. Res. 121, 1323–1330 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Correia, C. et al. Distinct carbon sources affect structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Sci. Rep. 7, 8590 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Yang, X. et al. Fatty acids enhance the maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cell Rep. 13, 657–668 (2019).

    Article  CAS  Google Scholar 

  49. Paredes, A. et al. γ-Linolenic acid in maternal milk drives cardiac metabolic maturation. Nature 618, 365–373 (2023).

    Article  CAS  PubMed  Google Scholar 

  50. Peters, M. C. et al. Metabolic maturation increases susceptibility to hypoxia-induced damage in human iPSC-derived cardiomyocytes. Stem Cells Transl. Med. 11, 1040–1051 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Haase, A. et al. GMP-compatible manufacturing of three iPS cell lines from human peripheral blood. Stem Cell Res. 35, 101394 (2019).

    Article  CAS  PubMed  Google Scholar 

  52. Papoutsakis, E. T. Media additives for protecting freely suspended animal cells against agitation and aeration damage. Trends Biotechnol. 9, 316–324 (1991).

    Article  CAS  PubMed  Google Scholar 

  53. Zhang, Z., Al-Rubeai, M. & Thomas, C. R. Effect of Pluronic F-68 on the mechanical properties of mammalian cells. Enzym. Microb. Technol. 14, 980–983 (1992).

    Article  CAS  Google Scholar 

  54. Haase, A., Göhring, G. & Martin, U. Generation of non-transgenic iPS cells from human cord blood CD34+ cells under animal component-free conditions. Stem Cell Res. 21, 71–73 (2017).

    Article  CAS  PubMed  Google Scholar 

  55. Haase, A. et al. Establishment of MHHi001-A-5, a GCaMP6f and RedStar dual reporter human iPSC line for in vitro and in vivo characterization and in situ tracing of iPSC derivatives. Stem Cell Res. 52, 102206 (2021).

    Article  CAS  PubMed  Google Scholar 

  56. Drakhlis, L., Devadas, S. B. & Zweigerdt, R. Generation of heart-forming organoids from human pluripotent stem cells. Nat. Protoc. 16, 5652–5672 (2021).

    Article  CAS  PubMed  Google Scholar 

  57. Chen, G. et al. Chemically defined conditions for human iPSC derivation and culture. Nat. Methods 8, 424–429 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Zhu, W.-Z., Van Biber, B. & Laflamme, M. A. Methods for the derivation and use of cardiomyocytes from human pluripotent stem cells. Methods Mol. Biol. 767, 419–431 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Drakhlis, L. et al. Human heart-forming organoids recapitulate early heart and foregut development. Nat. Biotechnol. 39, 737–746 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Cambier, L., Plate, M., Sucov, H. M. & Pashmforoush, M. Nkx2-5 regulates cardiac growth through modulation of Wnt signaling by R-spondin3. Development 141, 2959–2971 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Tarbit, E., Singh, I., Peart, J. N. & Rose’Meyer, R. B. Biomarkers for the identification of cardiac fibroblast and myofibroblast cells. Heart Fail. Rev. 24, 1–15 (2019).

    Article  CAS  PubMed  Google Scholar 

  62. Novak, D. et al. SOX2 in development and cancer biology. Semin. Cancer Biol. 67, 74–82 (2020).

    Article  CAS  PubMed  Google Scholar 

  63. Lu, H., Ma, J., Yang, Y., Shi, W. & Luo, L. EpCAM is an endoderm-specific Wnt derepressor that licenses hepatic development. Dev. Cell 24, 543–553 (2013).

    Article  CAS  PubMed  Google Scholar 

  64. Lertkiatmongkol, P., Liao, D., Mei, H., Hu, Y. & Newman, P. J. Endothelial functions of platelet/endothelial cell adhesion molecule-1 (CD31). Curr. Opin. Hematol. 23, 253–259 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Hamad, S. et al. High-efficient serum-free differentiation of endothelial cells from human iPS cells. Stem Cell Res. Ther. 13, 251 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the German Research Foundation (DFG; grants Cluster of Excellence REBIRTH EXC 62/2 and ZW64/4-2), the Federal Ministry of Education and Research (BMBF; grants 01EK1601A, 13XP5092B, 031L0249 and 01EK2108A), Lower Saxony ‘Förderung aus Mitteln des Niedersächsischen Vorab’ (grant ZN3340) and ‘Niedersächsische Ministerium für Wissenschaft und Kultur’ (MWK; grant ZN4092) and the European Union (Horizon Europe project HEAL grant 101056712). The views and opinions expressed are, however, those of the authors only and do not necessarily reflect those of the European Union or the European Health and Digital Executive Agency (HADEA). Neither the European Union nor the granting authority can be held responsible for them. We thank R. Bauerfeind and O. Terwolbeck from the MHH Core Unit for laser microscopy and for help with confocal microscopy.

Author information

Authors and Affiliations

Authors

Contributions

N.K., W.T., C.H., U.M. and R.Z. designed the experiments. N.K., W.T., C.H., M.M., A.F., L.D. and J.T. contributed to the experimental design, performed the experiments and analyzed the data. A.H. generated hiPSC lines. F.M., K.U. and C.H. developed scripts for automatized analysis of the experiments. N.K., W.T., U.M. and R.Z. wrote and reviewed the manuscript.

Corresponding authors

Correspondence to Nils Kriedemann or Robert Zweigerdt.

Ethics declarations

Competing interests

C.H. is an employee of Novo Nordisk. F.M. and W.T. are employees of Evotec. The other authors declare no competing interests.

Peer review

Peer review information

Nature Protocols thanks Kurt Pfannkuche 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 reference using this protocol

Halloin, C. et al. Stem Cell Rep. 13, 366–379 (2019): https://doi.org/10.1016/j.stemcr.2019.09.001

Extended data

Extended Data Fig. 1 Exemplary production of hiPSC-derived cardiomyocytes from vitronectin and (GMP-conforming) CTS vitronectin pre-culture.

a and b, Appearance of cells on CTS vitronectin (a) and vitronectin (b) on day −2 of the production protocol in monolayer (scale bar, 500 µm). c, Expression of markers of an undifferentiated state at CP 0 before induction of differentiation in samples from spinner flasks pre-cultured on vitronectin or CTS vitronectin. d, Cell yield at CP I of respective spinner-flask runs. e, Aggregate diameter of representative samples taken at CP I from respective spinners. Shown are individual values and mean ± s.d. in red. f, Percentage of cells positive for cardiac markers in representative samples from the respective spinner flasks at CP I.

Extended Data Fig. 2 Setup of the spinner flask.

In a sterile environment, unpack from plastic and install the lid with the stirrer. Remove one of the lids for the side ports and install a sampling device (optional).

Extended Data Fig. 3 Aggregate diameter size distribution for one differentiation run at CP 0, CP I and CP II.

Shown are individual aggregate diameters (gray) and mean values ± s.d. (red). The hiPSC line used is Amber.

Extended Data Fig. 4 Total aggregate count per milliliter of spinner flask experiments at CP 0 (n = 3) and CP I (n = 2).

Aggregate numbers were determined through dilution of a culture sample and manual counting through microscopy.

Extended Data Fig. 5 Confocal microscopy of aggregates at CP 0 and CP I to analyze cell number per aggregate.

a–c, Light microscope pictures of a representative cell sample at CP 0 (scale bar, 500 µm) (a) and confocal images of the central Z-stack of aggregates of the same batch (scale bar, 50 µm) (b and c). d–f, Light microscope pictures of a representative cell sample at CP I (scale bar, 500 µm) (d) and confocal images in a differentiated state as CM aggregates at CP I (scale bar, 50 µm) (e and f). Nuclei were stained with Sytox red after fixation; afterwards, aggregates were dehydrated with ethanol and cleared with methyl salicylate/benzyl benzoate (MSBB). Notable are pronounced cavities at CP 0 as well as at CP I. g, Aggregates treated for confocal imaging do not differ in diameter compared to untreated, equivalent samples measured according to QC3 at CP 0 or CP I. h, Nuclei identified at CP 0 and CP I through automatic counting of confocal z-stacks imaging whole aggregates. This nuclei count should closely resemble the total cell count per aggregate. i and j, Identified nuclei per aggregate in comparison to the individual aggregate diameter for aggregates at CP 0 (i) and CP I (j). Linear regression was added to depict the goodness of fit (CP 0 R2= 0.73; CP I R2= 0.55). For examples, see supplementary file z-stacks for CP 0 and CP I (open with the hyperstack function of FIJI/ImageJ). PFA, paraformaldehyde. The hiPSC line Phoenix was used in all experiments.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2

Supplementary Video 1

CM harvest for downstream application

Supplementary Video 2

hPSC splitting

Supplementary Video 3

Spinner medium exchange

Supplementary Data 1

Confocal z-stack pluripotent aggregates CP 0

Supplementary Data 2

Confocal z-stack CM aggregates CP I

Supplementary Code 1

ImageJ macro for aggregate size determination

Supplementary Code 2

ImageJ macro for viability score determination

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kriedemann, N., Triebert, W., Teske, J. et al. Standardized production of hPSC-derived cardiomyocyte aggregates in stirred spinner flasks. Nat Protoc (2024). https://doi.org/10.1038/s41596-024-00976-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41596-024-00976-2

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