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.

  • Article
  • Published:

Flow-enhanced vascularization and maturation of kidney organoids in vitro

Abstract

Kidney organoids derived from human pluripotent stem cells have glomerular- and tubular-like compartments that are largely avascular and immature in static culture. Here we report an in vitro method for culturing kidney organoids under flow on millifluidic chips, which expands their endogenous pool of endothelial progenitor cells and generates vascular networks with perfusable lumens surrounded by mural cells. We found that vascularized kidney organoids cultured under flow had more mature podocyte and tubular compartments with enhanced cellular polarity and adult gene expression compared with that in static controls. Glomerular vascular development progressed through intermediate stages akin to those involved in the embryonic mammalian kidney’s formation of capillary loops abutting foot processes. The association of vessels with these compartments was reduced after disruption of the endogenous VEGF gradient. The ability to induce substantial vascularization and morphological maturation of kidney organoids in vitro under flow opens new avenues for studies of kidney development, disease, and regeneration.

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: Developing kidney organoids cultured in vitro under high fluid flow exhibit enhanced vascularization during nephrogenesis.
Fig. 2: Intra- and interorganoid vascular networks with perfusable lumens supported by mural cells are observed for kidney organoids cultured under high flow in vitro.
Fig. 3: Tubular epithelia mature and undergo morphogenesis to become a polarized, ciliated compartment in contact with vasculature in response to high-flow conditions on chip.
Fig. 4: Flow-enhanced glomerular vascularization and morphogenesis of kidney organoids in vitro mirrors stages of glomerular development in vivo.

Similar content being viewed by others

Data availability

The data generated in this study are available from the corresponding authors upon request.

References

  1. Morizane, R. & Bonventre, J. V. Generation of nephron progenitor cells and kidney organoids from human pluripotent stem cells. Nat. Protoc. 12, 195–207 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Morizane, R. et al. Nephron organoids derived from human pluripotent stem cells model kidney development and injury. Nat. Biotechnol. 33, 1193–1200 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Takasato, M. et al. Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature 526, 564–568 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Lam, A. Q. et al. Rapid and efficient differentiation of human pluripotent stem cells into intermediate mesoderm that forms tubules expressing kidney proximal tubular markers. J. Am. Soc. Nephrol. 25, 1211–1225 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Freedman, B. S. et al. Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nat. Commun. 6, 8715 (2015).

    Article  CAS  PubMed  Google Scholar 

  6. Morizane, R. & Bonventre, J. V. Kidney organoids: a translational journey. Trends Mol. Med. 23, 246–263 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Taguchi, A. et al. Redefining the in vivo origin of metanephric nephron progenitors enables generation of complex kidney structures from pluripotent stem cells. Cell Stem Cell 14, 53–67 (2014).

    Article  CAS  PubMed  Google Scholar 

  8. Takasato, M. & Little, M. H. A strategy for generating kidney organoids: recapitulating the development in human pluripotent stem cells. Dev. Biol. 420, 210–220 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wu, H. et al. Comparative analysis of kidney organoid and adult human kidney single cell and single nucleus transcriptomes. bioRxiv Preprint at https://www.biorxiv.org/content/early/2017/12/11/232561 (2017).

  10. van den Berg, C. W. et al. Renal subcapsular transplantation of PSC-derived kidney organoids induces neo-vasculogenesis and significant glomerular and tubular maturation in vivo. Stem Cell Rep. 10, 751–765 (2018).

    Article  Google Scholar 

  11. Takebe, T. et al. Vascularized and complex organ buds from diverse tissues via mesenchymal cell-driven condensation. Cell Stem Cell 16, 556–565 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Bantounas, I. et al. Generation of functioning nephrons by implanting human pluripotent stem cell-derived kidney progenitors. Stem Cell Rep. 10, 766–779 (2018).

    Article  Google Scholar 

  13. Camp, J. G. et al. Multilineage communication regulates human liver bud development from pluripotency. Nature 546, 533–538 (2017).

    CAS  PubMed  Google Scholar 

  14. Kolesky, D. B. et al. 3D bioprinting of vascularized, heterogeneous cell-laden tissue constructs. Adv. Mater. 26, 3124–3130 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Homan, K. A. et al. Bioprinting of 3D convoluted renal proximal tubules on perfusable chips. Sci. Rep. 6, 34845 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Halt, K. J. et al. CD146+ cells are essential for kidney vasculature development. Kidney Int. 90, 311–324 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Zudaire, E., Gambardella, L., Kurcz, C. & Vermeren, S. A computational tool for quantitative analysis of vascular networks. PLoS One 6, e27385 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Munro, D. A. D., Hohenstein, P. & Davies, J. A. Cycles of vascular plexus formation within the nephrogenic zone of the developing mouse kidney. Sci. Rep. 7, 3273 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Daniel, E. et al. Spatiotemporal heterogeneity and patterning of developing renal blood vessels. Angiogenesis 21, 617–634 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Robert, B., St John, P. L. & Abrahamson, D. R. Direct visualization of renal vascular morphogenesis in Flk1 heterozygous mutant mice. Am. J. Physiol. 275, F164–F172 (1998).

    CAS  PubMed  Google Scholar 

  21. McMahon, A. P. Development of the mammalian kidney. Curr. Top. Dev. Biol. 117, 31–64 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Abrahamson, D. R. Development of kidney glomerular endothelial cells and their role in basement membrane assembly. Organogenesis 5, 275–287 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Scheppke, L. et al. Notch promotes vascular maturation by inducing integrin-mediated smooth muscle cell adhesion to the endothelial basement membrane. Blood 119, 2149–2158 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wu, S., Kim, C., Baer, L. & Zhu, X. Bevacizumab increases risk for severe proteinuria in cancer patients. J. Am. Soc. Nephrol. 21, 1381–1389 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sharmin, S. et al. Human induced pluripotent stem cell-derived podocytes mature into vascularized glomeruli upon experimental transplantation. J. Am. Soc. Nephrol. 27, 1778–1791 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Serluca, F. C., Drummond, I. A. & Fishman, M. C. Endothelial signaling in kidney morphogenesis: a role for hemodynamic forces. Curr. Biol. 12, 492–497 (2002).

    Article  CAS  PubMed  Google Scholar 

  27. Ichimura, K. et al. Morphological process of podocyte development revealed by block-face scanning electron microscopy. J. Cell. Sci. 130, 132–142 (2017).

    Article  CAS  PubMed  Google Scholar 

  28. Huch, M., Knoblich, J. A., Lutolf, M. P. & Martinez-Arias, A. The hope and the hype of organoid research. Development 144, 938–941 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Oxburgh, L. & Carroll, T. J. The bioengineered kidney: science or science fiction? Curr. Opin. Nephrol. Hypertens. 25, 343–347 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Little, M. H. Growing kidney tissue from stem cells: how far from “party trick” to medical application? Cell Stem Cell 18, 695–698 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ainslie, K. M., Garanich, J. S., Dull, R. O. & Tarbell, J. M. Vascular smooth muscle cell glycocalyx influences shear stress-mediated contractile response. J. Appl. Physiol. 98, 242–249 (2005).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank P. Galichon for flow cytometry analyses; Y. Yoda and K. Susa for cell culture and immunocytochemistry; S. Jain at The Washington University Kidney Translational Research Center (KTRC; St. Louis, MO, USA) for providing the BJFF hiPSC line; A. Moisan, C. Chen, and S. Uzel for insightful discussions; J. Weaver, B. Roman-Manso, N. Zhou, and M. Ericsson for imaging assistance; and L. Sanders for videography. This study was supported by the US National Institutes of Health (NIH; T32 fellowship training grant DK007527 to N.G.; Subaward U01DK107350 to M.T.V.; R37 grant DK039773 to J.V.B.; UG3 grant TR002155 to J.V.B., M.T.V., J.A.L., and R.M.; grant P30 DK079333 (the BJFF line) supporting The Washington University KTRC), the Harvard Stem Cell Institute (interdisciplinary grant to N.G.; seed grant to R.M. and J.A.L.), Brigham and Women’s Hospital (Research Excellence Award to N.G. and R.M.; Faculty Career Development Award to R.M.), the NIDDK Diabetic Complications Consortium (DiaComp, https://www.diacomp.org; grant DK076169 to R.M.), the NIH (Re)Building a Kidney Consortium (U01DK107350 to K.A.H. and J.A.L.), the Office of Naval Research Vannevar Bush Faculty Fellowship program (award no. N000141612823 to M.S.-S. and J.A.L.), and the Wyss Institute for Biologically Inspired Engineering (D.B.K., K.T.K., D.M., and J.A.L.). J.A.L. thanks the GETTYLAB and S. Lindenfeld for their generous donations in support of this research. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

K.A.H., N.G., J.A.L., R.M., D.B.K., and J.V.B. conceived the project. K.A.H., N.G., and K.T.K. designed the research, and R.M. and J.A.L. supervised the research. K.A.H., N.G., K.T.K., R.M., and D.B.K. designed, performed, and analyzed all experiments. M.T.V. provided critical insights into embryonic development, cell sources, and mouse embryonic kidneys. M.S.-S. designed and built the silicone millifluidic chips, interfacing with perfusion pumps, and analyzed the fluid flow profiles on chip. D.M. and T.M. sourced and validated antibodies, optimized staining protocols, and provided invaluable cell culture analysis and support. T.F. developed methodology for quantifying vascular and tubule features in confocal imaging stacks. All authors contributed to manuscript writing.

Corresponding authors

Correspondence to Jennifer A. Lewis or Ryuji Morizane.

Ethics declarations

Competing interests

J.V.B. and R.M. are co-inventors on patents (PCT/US16/52350) on organoid technologies that are assigned to Partners Healthcare. J.V.B. or his family has received income for consulting from companies interested in biomarkers: Sekisui, Millennium, Johnson & Johnson, and Novartis. J.V.B. is a co-founder of, is a consultant to, and owns equity in Goldfinch Bio. K.A.H. is a co-founder and chairwoman of NanoHybrids Inc. J.A.L. is a co-founder of and owns equity in Voxel8 Inc.

Additional information

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

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–22 and Supplementary Tables 1–3

Reporting Summary

Supplementary Video 1

Vascular precursors and mature vessels in a kidney organoid cultured under high -flow conditions. Confocal z-stack movie of a whole organoid, containing multiple PODXL+ cellular clusters, permeated by PECAM1+ vessels supported by MCAM+ cells. DAPI (blue), 4’,6-diamidino-2-phenylindole; PECAM1 (red), CD31; MCAM (yellow), CD146; PODXL (cyan), podocalyxin.

Supplementary Video 2

Bead perfusion at the bottom edge of a live organoid. Beads (green; 100 nm) were perfused in media over the top of an organoid in high-flow conditions. The beads accumulate with time at the edge (white dotted line). ULEX staining (red) was applied to the media and highlights the vessels. Halfway through the movie, the red channel laser is turned off in order to visualize the beads in the green channel only. As is common with this system, one vessel is perfused and a nearby vessel is not, showing that perfusion in lumens in this system is happening, but not evenly throughout the vascular tree. Scale bar, 10 μm.

Supplementary Video 3

Kidney organoid fusion under flow. Bright-field time-lapse movie of two organoids fusing after being seeded on chip under high flow during a 47-h period (images taken every 2 min). (Note: the period shown corresponds to days 1–3 under perfusion.)

Supplementary Video 4

Vascular–tubule interactions in kidney organoids under high flow. PECAM1+ cells (red) both wrap and extend longitudinally around tubules. DAPI (blue), 4’,6-diamidino-2-phenylindole; PECAM1 (red), CD31; PODXL (cyan), podocalyxin.

Supplementary Video 5

Capillary invasion into S-shaped bodies and PODXL+ lobules in kidney organoids cultured under high flow. Z-stack confocal slices through various glomeruli in organoids under high-flow conditions (z-stacks of the still images in Fig. 4d,e in the text).

Supplementary Video 6

Confocal z-stack movies of PODXL+ lobules with PECAM1+ vessels supported by MCAM+ cells for kidney organoids cultured under high flow at day 21. DAPI (blue), 4’,6-diamidino-2-phenylindole; PECAM1 (red), CD31; MCAM (yellow), CD146; PODXL (cyan), podocalyxin.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Homan, K.A., Gupta, N., Kroll, K.T. et al. Flow-enhanced vascularization and maturation of kidney organoids in vitro. Nat Methods 16, 255–262 (2019). https://doi.org/10.1038/s41592-019-0325-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-019-0325-y

This article is cited by

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