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:

Generation of blood vessel organoids from human pluripotent stem cells

Abstract

Blood vessels are fundamental to animal life and have critical roles in many diseases, such as stroke, myocardial infarction and diabetes. The vasculature is formed by endothelial cells that line the vessel and are covered with mural cells, specifically pericytes in smaller vessels and vascular smooth muscle cells (vSMCs) in larger-diameter vessels. Both endothelial cells and mural cells are essential for proper blood vessel function and can be derived from human pluripotent stem cells (hPSCs). Here, we describe a protocol to generate self-organizing 3D human blood vessel organoids from hPSCs that exhibit morphological, functional and molecular features of human microvasculature. These organoids are differentiated via mesoderm induction of hPSC aggregates and subsequent differentiation into endothelial networks and pericytes in a 3D collagen I–Matrigel matrix. Blood vessels form within 2–3 weeks and can be further grown in scalable suspension culture. Importantly, in vitro–differentiated human blood vessel organoids transplanted into immunocompromised mice gain access to the mouse circulation and specify into functional arteries, arterioles and veins.

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 diagram of the time line for generating blood vessel organoids from hPSCs.
Fig. 2: Bright-field images of hPSC differentiation into vascular networks and blood vessel organoids.
Fig. 3: Characterization of vascular networks and blood vessel organoids differentiated from hPSCs.
Fig. 4: Characterization of blood vessel organoids transplanted into NSG mice.

Similar content being viewed by others

Data availability

The data generated or analyzed during this study are included in this published article.

References

  1. Carmeliet, P. Angiogenesis in health and disease. Nat. Med. 9, 653–660 (2003).

    Article  CAS  Google Scholar 

  2. Park, C. A hierarchical order of factors in the generation of FLK1- and SCL-expressing hematopoietic and endothelial progenitors from embryonic stem cells. Development 131, 2749–2762 (2004).

    Article  CAS  Google Scholar 

  3. Faloon, P. et al. Basic fibroblast growth factor positively regulates hematopoietic development. Development 127, 1931–1941 (2000).

    CAS  PubMed  Google Scholar 

  4. Ferguson, J. E., Kelley, R. W. & Patterson, C. Mechanisms of endothelial differentiation in embryonic vasculogenesis. Arterioscler. Thromb. Vasc. Biol. 25, 2246–2256 (2005).

    Article  CAS  Google Scholar 

  5. Potente, M., Gerhardt, H. & Carmeliet, P. Basic and therapeutic aspects of angiogenesis. Cell 146, 873–887 (2011).

    Article  CAS  Google Scholar 

  6. Gebala, V., Collins, R., Geudens, I., Phng, L. K. & Gerhardt, H. Blood flow drives lumen formation by inverse membrane blebbing during angiogenesis in vivo. Nat. Cell Biol. 18, 443–450 (2016).

    Article  CAS  Google Scholar 

  7. Charpentier, M. S. & Conlon, F. L. Cellular and molecular mechanisms underlying blood vessel lumen formation. BioEssays 36, 251–259 (2014).

    Article  CAS  Google Scholar 

  8. Simons, M., Gordon, E. & Claesson-Welsh, L. Mechanisms and regulation of endothelial VEGF receptor signalling. Nat. Rev. Mol. Cell Biol. 17, 611–625 (2016).

    Article  CAS  Google Scholar 

  9. Armulik, A., Genové, G. & Betsholtz, C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev. Cell 21, 193–215 (2011).

    Article  CAS  Google Scholar 

  10. Yamashita, J. et al. Flk1-positive cells derived from embryonic stem cells serve as vascular progenitors. Nature 408, 92–96 (2000).

    Article  CAS  Google Scholar 

  11. Gerhardt, H. et al. VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. J. Cell Biol. 161, 1163–1177 (2003).

    Article  CAS  Google Scholar 

  12. Stratman, A. N. & Davis, G. E. Endothelial cell-pericyte interactions stimulate basement membrane matrix assembly: influence on vascular tube remodeling, maturation, and stabilization. Microsc. Microanal. 18, 68–80 (2012).

    Article  CAS  Google Scholar 

  13. Lindahl, P., Johansson, B. R., Levéen, P. & Betsholtz, C. Pericyte loss and microaneurysm formation in PDGF-B-deficient mice. Science 277, 242–245 (1997).

    Article  CAS  Google Scholar 

  14. Davis, G. E., Norden, P. R. & Bowers, S. L. K. Molecular control of capillary morphogenesis and maturation by recognition and remodeling of the extracellular matrix: functional roles of endothelial cells and pericytes in health and disease. Connect. Tissue Res. 56, 392–402 (2015).

    Article  Google Scholar 

  15. Wimmer, R. A. et al. Human blood vessel organoids as a model of diabetic vasculopathy. Nature 565, 505–510 (2019).

    Article  CAS  Google Scholar 

  16. Adams, W. J. et al. Functional vascular endothelium derived from human induced pluripotent stem cells. Stem Cell Rep. 1, 105–113 (2013).

    Article  CAS  Google Scholar 

  17. Rufaihah, A. J. et al. Human induced pluripotent stem cell-derived endothelial cells exhibit functional heterogeneity. Am. J. Transl. Res. 5, 21–35 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Kusuma, S. et al. Self-organized vascular networks from human pluripotent stem cells in a synthetic matrix. Proc. Natl. Acad. Sci. USA 110, 12601–12606 (2013).

    Article  CAS  Google Scholar 

  19. Orlova, V. V. et al. Generation, expansion and functional analysis of endothelial cells and pericytes derived from human pluripotent stem cells. Nat. Protoc. 9, 1514–1531 (2014).

    Article  CAS  Google Scholar 

  20. Orlova, V. V. et al. Functionality of endothelial cells and pericytes from human pluripotent stem cells demonstrated in cultured vascular plexus and zebrafish xenografts. Arterioscler. Thromb. Vasc. Biol. 34, 177–186 (2014).

    Article  CAS  Google Scholar 

  21. Lian, X. et al. Efficient differentiation of human pluripotent stem cells to endothelial progenitors via small-molecule activation of WNT signaling. Stem Cell Rep. 3, 804–816 (2014).

    Article  CAS  Google Scholar 

  22. Cheung, C., Bernardo, A. S., Trotter, M. W. B., Pedersen, R. A. & Sinha, S. Generation of human vascular smooth muscle subtypes provides insight into embryological origing-dependent disease susceptibility. Nat. Biotechnol. 30, 165–173 (2012).

    Article  CAS  Google Scholar 

  23. Bajpai, V. K., Mistriotis, P., Loh, Y. H., Daley, G. Q. & Andreadis, S. T. Functional vascular smooth muscle cells derived from human induced pluripotent stem cells via mesenchymal stem cell intermediates. Cardiovasc. Res 96, 391–400 (2012).

    Article  CAS  Google Scholar 

  24. Trotter, M. W. B., Cheung, C., Pedersen, R. A., Sinha, S. & Bernardo, A. S. Generation of human vascular smooth muscle subtypes provides insight into embryological origin–dependent disease susceptibility. Nat. Biotechnol. 30, 165–173 (2012).

    Article  Google Scholar 

  25. Ren, X. et al. Engineering pulmonary vasculature in decellularized rat and human lungs. Nat. Biotechnol. 33, 1097–1102 (2015).

    Article  CAS  Google Scholar 

  26. Samuel, R. et al. Generation of functionally competent and durable engineered blood vessels from human induced pluripotent stem cells. Proc. Natl. Acad. Sci. USA 110, 12774–12779 (2013).

    Article  CAS  Google Scholar 

  27. James, D. et al. Expansion and maintenance of human embryonic stem cell-derived endothelial cells by TGFΒ inhibition is Id1 dependent. Nat. Biotechnol. 28, 161–166 (2010).

    Article  CAS  Google Scholar 

  28. Patsch, C. et al. Generation of vascular endothelial and smooth muscle cells from human pluripotent stem cells. Nat. Cell Biol. 17, 994–1003 (2015).

    Article  CAS  Google Scholar 

  29. Samuel, R., Duda, D. G., Fukumura, D. & Jain, R. K. Vascular diseases await translation of blood vessels engineered from stem cells. Sci. Transl. Med. 7, 309rv6 (2015).

    Article  Google Scholar 

  30. Hockemeyer, D. & Jaenisch, R. Induced pluripotent stem cells meet genome editing. Cell Stem Cell 18, 573–586 (2016).

    Article  CAS  Google Scholar 

  31. Potente, M. & Mäkinen, T. Vascular heterogeneity and specialization in development and disease. Nat. Rev. Mol. Cell Biol. 18, 477–494 (2017).

    Article  CAS  Google Scholar 

  32. Homan, K. A. et al. Flow-enhanced vascularization and maturation of kidney organoids in vitro. Nat. Methods 16, 255–262 (2019).

    Article  CAS  Google Scholar 

  33. Augustin, H. G. & Koh, G. Y. Organotypic vasculature: from descriptive heterogeneity to functional pathophysiology. Science 357, eaal2379 (2017).

    Article  Google Scholar 

  34. Ungrin, M. D., Joshi, C., Nica, A., Bauwens, C. & Zandstra, P. W. Reproducible, ultra high-throughput formation of multicellular organization from single cell suspension-derived human embryonic stem cell aggregates. PLoS ONE 3, e1565 (2008).

    Article  Google Scholar 

  35. Huttala, O. et al. Human vascular model with defined stimulation medium—a characterization study. ALTEX 32, 125–136 (2015).

    Article  Google Scholar 

  36. Gjorevski, N. et al. Designer matrices for intestinal stem cell and organoid culture. Nature 539, 560–564 (2016).

    Article  CAS  Google Scholar 

  37. Takebe, T. et al. Generation of a vascularized and functional human liver from an iPSC-derived organ bud transplant. Nat. Protoc. 9, 396–409 (2014).

    Article  CAS  Google Scholar 

  38. Beers, J. et al. Passaging and colony expansion of human pluripotent stem cells by enzyme-free dissociation in chemically defined culture conditions. Nat. Protoc. 7, 2029–2040 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank all members of our laboratories for constructive critiques and expert advice. We also thank M. Boehm (Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health) for sharing the iPSC line NC8 and A. Kavirayani, M. Zeba, T. Engelmaier, J. Klughofer and A. Piszczek for histology services. J.M.P. was supported by grants from IMBA, the Austrian Ministry of Sciences, the Austrian Academy of Sciences, an ERC grant and an Era of Hope Innovator award.

Author information

Authors and Affiliations

Authors

Contributions

R.A.W., A.L. and M.A. performed experiments. R.A.W., D.K. and J.M.P. supervised the project. R.A.W. and J.M.P. wrote the manuscript.

Corresponding authors

Correspondence to Reiner A. Wimmer or Josef M. Penninger.

Ethics declarations

Competing interests

A patent application related to this work has been filed. IMBA is in the process of applying for a patent application covering vascular organoid technology that lists R.A.W., D.K. and J.M.P. as inventors.

Additional information

Journal peer review information Nature Protocols thanks Andrew Baker and Valeria Orlova for their contributions 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 link

Key reference using this protocol

Wimmer, R.A. et al. Nature 565, 505–510 (2019): https://doi.org/10.1038/s41586-018-0858-8

Supplementary information

Supplementary Video 1

Dissection of vascular networks for blood vessel organoid production

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wimmer, R.A., Leopoldi, A., Aichinger, M. et al. Generation of blood vessel organoids from human pluripotent stem cells. Nat Protoc 14, 3082–3100 (2019). https://doi.org/10.1038/s41596-019-0213-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-019-0213-z

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