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:

Nephron organoids derived from human pluripotent stem cells model kidney development and injury

Abstract

Kidney cells and tissues derived from human pluripotent stem cells (hPSCs) may enable organ regeneration, disease modeling and drug screening. We report an efficient, chemically defined protocol for differentiating hPSCs into multipotent nephron progenitor cells (NPCs) that can form nephron-like structures. By recapitulating metanephric kidney development in vitro, we generate SIX2+SALL1+WT1+PAX2+ NPCs with 90% efficiency within 9 days of differentiation. The NPCs possess the developmental potential of their in vivo counterparts and form PAX8+LHX1+ renal vesicles that self-organize into nephron structures. In both two- and three-dimensional culture, NPCs form kidney organoids containing epithelial nephron-like structures expressing markers of podocytes, proximal tubules, loops of Henle and distal tubules in an organized, continuous arrangement that resembles the nephron in vivo. We also show that this organoid culture system can be used to study mechanisms of human kidney development and toxicity.

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

Figure 1: Differentiation of hPSCs into posterior intermediate mesoderm.
Figure 2: Differentiation into nephron progenitor cells and spontaneous formation of renal vesicles.
Figure 3: Induction of pretubular aggregates and renal vesicles from nephron progenitor cells.
Figure 4: Self-organizing nephron formation in 2D culture.
Figure 5: Self-organizing nephron formation in 3D culture.
Figure 6: Modeling kidney development and injury in kidney organoids.

Similar content being viewed by others

References

  1. Coresh, J. et al. Prevalence of chronic kidney disease in the United States. J. Am. Med. Assoc. 298, 2038–2047 (2007).

    Article  CAS  Google Scholar 

  2. Herzlinger, D., Koseki, C., Mikawa, T. & al-Awqati, Q. Metanephric mesenchyme contains multipotent stem cells whose fate is restricted after induction. Development 114, 565–572 (1992).

    CAS  PubMed  Google Scholar 

  3. Self, M. et al. Six2 is required for suppression of nephrogenesis and progenitor renewal in the developing kidney. EMBO J. 25, 5214–5228 (2006).

    Article  CAS  Google Scholar 

  4. Kobayashi, A. et al. Six2 defines and regulates a multipotent self-renewing nephron progenitor population throughout mammalian kidney development. Cell Stem Cell 3, 169–181 (2008).

    Article  CAS  Google Scholar 

  5. Boyle, S. et al. Fate mapping using Cited1-CreERT2 mice demonstrates that the cap mesenchyme contains self-renewing progenitor cells and gives rise exclusively to nephronic epithelia. Dev. Biol. 313, 234–245 (2008).

    Article  CAS  Google Scholar 

  6. Hinchliffe, S.A., Sargent, P.H., Howard, C.V., Chan, Y.F. & van Velzen, D. Human intrauterine renal growth expressed in absolute number of glomeruli assessed by the disector method and Cavalieri principle. Lab. Invest. 64, 777–784 (1991).

    CAS  PubMed  Google Scholar 

  7. Humphreys, B.D. et al. Intrinsic epithelial cells repair the kidney after injury. Cell Stem Cell 2, 284–291 (2008).

    Article  CAS  Google Scholar 

  8. Georgas, K.M., Chiu, H.S., Lesieur, E., Rumballe, B.A. & Little, M.H. Expression of metanephric nephron-patterning genes in differentiating mesonephric tubules. Dev. Dyn. 240, 1600–1612 (2011).

    Article  CAS  Google Scholar 

  9. Kim, D. & Dressler, G.R. Nephrogenic factors promote differentiation of mouse embryonic stem cells into renal epithelia. J. Am. Soc. Nephrol. 16, 3527–3534 (2005).

    Article  CAS  Google Scholar 

  10. Vigneau, C. et al. Mouse embryonic stem cell-derived embryoid bodies generate progenitors that integrate long term into renal proximal tubules in vivo. J. Am. Soc. Nephrol. 18, 1709–1720 (2007).

    Article  CAS  Google Scholar 

  11. Morizane, R., Monkawa, T. & Itoh, H. Differentiation of murine embryonic stem and induced pluripotent stem cells to renal lineage in vitro. Biochem. Biophys. Res. Commun. 390, 1334–1339 (2009).

    Article  CAS  Google Scholar 

  12. Narayanan, K. et al. Human embryonic stem cells differentiate into functional renal proximal tubular-like cells. Kidney Int. 83, 593–603 (2013).

    Article  CAS  Google Scholar 

  13. Morizane, R. et al. Kidney specific protein-positive cells derived from embryonic stem cells reproduce tubular structures in vitro and differentiate into renal tubular cells. PLoS One 8, e64843 (2014).

    Article  Google Scholar 

  14. Mae, S. et al. Monitoring and robust induction of nephrogenic intermediate mesoderm from human pluripotent stem cells. Nat. Commun. 4, 1367 (2013).

    Article  Google Scholar 

  15. Xia, Y. et al. Directed differentiation of human pluripotent cells to ureteric bud kidney progenitor-like cells. Nat. Cell Biol. 15, 1507–1515 (2013).

    Article  CAS  Google Scholar 

  16. Lam, A.Q., Freedman, B.S. & Bonventre, J.V. Directed differentiation of pluripotent stem cells to kidney cells. Semin. Nephrol. 34, 445–461 (2014).

    Article  CAS  Google Scholar 

  17. 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  Google Scholar 

  18. Takasato, M. et al. Directing human embryonic stem cell differentiation towards a renal lineage generates a self-organizing kidney. Nat. Cell Biol. 16, 118–126 (2014).

    Article  CAS  Google Scholar 

  19. Kang, M. & Han, Y.M. Differentiation of human pluripotent stem cells into nephron progenitor cells in a serum and feeder free system. PLoS One 9, e94888 (2014).

    Article  Google Scholar 

  20. 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  Google Scholar 

  21. Imberti, B. et al. Renal progenitors derived from human iPSCs engraft and restore function in a mouse model of acute kidney injury. Sci. Rep. 5, 8826 (2015).

    Article  Google Scholar 

  22. Grote, D., Souabni, A., Busslinger, M. & Bouchard, M. Pax 2/8-regulated Gata 3 expression is necessary for morphogenesis and guidance of the nephric duct in the developing kidney. Development 133, 53–61 (2006).

    Article  CAS  Google Scholar 

  23. Attia, L., Yelin, R. & Schultheiss, T.M. Analysis of nephric duct specification in the avian embryo. Development 139, 4143–4151 (2012).

    Article  CAS  Google Scholar 

  24. Lengerke, C. et al. BMP and Wnt specify hematopoietic fate by activation of the Cdx-Hox pathway. Cell Stem Cell 2, 72–82 (2008).

    Article  CAS  Google Scholar 

  25. Liu, P. et al. Requirement for Wnt3 in vertebrate axis formation. Nat. Genet. 22, 361–365 (1999).

    Article  CAS  Google Scholar 

  26. Iimura, T. & Pourquié, O. Collinear activation of Hoxb genes during gastrulation is linked to mesoderm cell ingression. Nature 442, 568–571 (2006).

    Article  CAS  Google Scholar 

  27. Deschamps, J. & van Nes, J. Developmental regulation of the Hox genes during axial morphogenesis in the mouse. Development 132, 2931–2942 (2005).

    Article  CAS  Google Scholar 

  28. Osafune, K. et al. Marked differences in differentiation propensity among human embryonic stem cell lines. Nat. Biotechnol. 26, 313–315 (2008).

    Article  CAS  Google Scholar 

  29. Bock, C. et al. Reference Maps of human ES and iPS cell variation enable high-throughput characterization of pluripotent cell lines. Cell 144, 439–452 (2011).

    Article  CAS  Google Scholar 

  30. Mahlapuu, M., Ormestad, M., Enerbäck, S. & Carlsson, P. The forkhead transcription factor Foxf1 is required for differentiation of extra-embryonic and lateral plate mesoderm. Development 128, 155–166 (2001).

    CAS  PubMed  Google Scholar 

  31. Zimmerman, L.B., De Jesús-Escobar, J.M. & Harland, R.M. The Spemann organizer signal noggin binds and inactivates bone morphogenetic protein 4. Cell 86, 599–606 (1996).

    Article  CAS  Google Scholar 

  32. Barak, H. et al. FGF9 and FGF20 maintain the stemness of nephron progenitors in mice and man. Dev. Cell 22, 1191–1207 (2012).

    Article  CAS  Google Scholar 

  33. Narlis, M., Grote, D., Gaitan, Y., Boualia, S.K. & Bouchard, M. Pax2 and pax8 regulate branching morphogenesis and nephron differentiation in the developing kidney. J. Am. Soc. Nephrol. 18, 1121–1129 (2007).

    Article  CAS  Google Scholar 

  34. Kispert, A., Vainio, S. & McMahon, A.P. Wnt-4 is a mesenchymal signal for epithelial transformation of metanephric mesenchyme in the developing kidney. Development 125, 4225–4234 (1998).

    CAS  PubMed  Google Scholar 

  35. Barasch, J., Pressler, L., Connor, J. & Malik, A. A ureteric bud cell line induces nephrogenesis in two steps by two distinct signals. Am. J. Physiol. 271, F50–F61 (1996).

    CAS  PubMed  Google Scholar 

  36. Carroll, T.J., Park, J.S., Hayashi, S., Majumdar, A. & McMahon, A.P. Wnt9b plays a central role in the regulation of mesenchymal to epithelial transitions underlying organogenesis of the mammalian urogenital system. Dev. Cell 9, 283–292 (2005).

    Article  CAS  Google Scholar 

  37. Potter, S.S., Hartman, H.A., Kwan, K.M., Behringer, R.R. & Patterson, L.T. Laser capture-microarray analysis of Lim1 mutant kidney development. Genesis 45, 432–439 (2007).

    Article  CAS  Google Scholar 

  38. Park, J.S. et al. Six2 and Wnt regulate self-renewal and commitment of nephron progenitors through shared gene regulatory networks. Dev. Cell 23, 637–651 (2012).

    Article  CAS  Google Scholar 

  39. Nouwen, E.J., Dauwe, S., van der Biest, I. & De Broe, M.E. Stage- and segment-specific expression of cell-adhesion molecules N-CAM, A-CAM, and L-CAM in the kidney. Kidney Int. 44, 147–158 (1993).

    Article  CAS  Google Scholar 

  40. Nakai, S. et al. Crucial roles of Brn1 in distal tubule formation and function in mouse kidney. Development 130, 4751–4759 (2003).

    Article  CAS  Google Scholar 

  41. Georgas, K. et al. Use of dual section mRNA in situ hybridisation/immunohistochemistry to clarify gene expression patterns during the early stages of nephron development in the embryo and in the mature nephron of the adult mouse kidney. Histochem. Cell Biol. 130, 927–942 (2008).

    Article  CAS  Google Scholar 

  42. Georgas, K. et al. Analysis of early nephron patterning reveals a role for distal RV proliferation in fusion to the ureteric tip via a cap mesenchyme-derived connecting segment. Dev. Biol. 332, 273–286 (2009).

    Article  CAS  Google Scholar 

  43. Cheng, H.T. et al. Gamma-secretase activity is dispensable for mesenchyme-to-epithelium transition but required for podocyte and proximal tubule formation in developing mouse kidney. Development 130, 5031–5042 (2003).

    Article  CAS  Google Scholar 

  44. Cheng, H.T. et al. Notch2, but not Notch1, is required for proximal fate acquisition in the mammalian nephron. Development 134, 801–811 (2007).

    Article  CAS  Google Scholar 

  45. Uchino, S. et al. Acute renal failure in critically ill patients: a multinational, multicenter study. J. Am. Med. Assoc. 294, 813–818 (2005).

    Article  CAS  Google Scholar 

  46. Whiting, P.H. & Brown, P.A. The relationship between enzymuria and kidney enzyme activities in experimental gentamicin nephrotoxicity. Ren. Fail. 18, 899–909 (1996).

    Article  CAS  Google Scholar 

  47. Vaidya, V.S. et al. Kidney injury molecule-1 outperforms traditional biomarkers of kidney injury in preclinical biomarker qualification studies. Nat. Biotechnol. 28, 478–485 (2010).

    Article  CAS  Google Scholar 

  48. Taguchi, A. & Nishinakamura, R. Nephron reconstitution from pluripotent stem cells. Kidney Int. 87, 894–900 (2015).

    Article  CAS  Google Scholar 

  49. Lancaster, M.A. et al. Cerebral organoids model human brain development and microcephaly. Nature 501, 373–379 (2013).

    Article  CAS  Google Scholar 

  50. McCracken, K.W. et al. Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature 516, 400–404 (2014).

    Article  CAS  Google Scholar 

  51. Freedman, B.S. et al. Reduced ciliary polycystin-2 in induced pluripotent stem cells from polycystic kidney disease patients with PKD1 mutations. J. Am. Soc. Nephrol. 24, 1571–1586 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank V. Bijol for providing electron microscopy images of normal human kidneys, L. Racusen (Johns Hopkins Hospital) for HKC-8, J. Barasch (Columbia University) for a mouse ureteric bud cell line, and A.P. McMahon (University of Southern California) for NIH3T3-Wnt4. This study was supported by US National Institutes of Health (NIH) grants R37 DK039773 and R01 DK072381 (J.V.B.); Grant-in-Aid for JSPS (Japan Society for the Promotion of Science); Postdoctoral Fellowship for Research Abroad (R.M.); American Heart Association grant 11FTF7320023 (A.Q.L.); Harvard Stem Cell Institute (A.Q.L., J.V.B. and M.T.V.); and NIH DK102826 and National Kidney Foundation Young Investigator Grant (B.S.F.).

Author information

Authors and Affiliations

Authors

Contributions

R.M. and J.V.B. formulated the strategy for this study. R.M. designed and performed experiments. R.M., A.Q.L. and J.V.B. wrote the manuscript. A.Q.L. and B.S.F. performed nephrotoxicity assays. S.K. performed real-time PCR. M.T.V. and J.V.B. helped to design experiments. All authors helped to interpret the results.

Corresponding authors

Correspondence to Ryuji Morizane or Joseph V Bonventre.

Ethics declarations

Competing interests

J.V.B. is a co-inventor on KIM-1 patents, which have been licensed by Partners Healthcare to several companies. He has received royalty income from Partners Healthcare. J.V.B. or his family has received income for consulting from companies interested in biomarkers: Sekisui, Millennium, Johnson & Johnson and Novartis.

Integrated supplementary information

Supplementary Figure 1 Metanephric development and published protocols

(a) A schematic illustration of intermediate mesoderm and subsequent differentiation into mesonephros and metanephros. (b) The summary and comparison of published protocols and our new protocol. Takasato et al. Nat Cell Biol. 2014. Taguchi et al. Cell Stem Cell. 2014. RA: retinoic acid.

Supplementary Figure 2 Adjustment of the dose and CHIR treatment time

(a) A schematic illustration of primitive streak and subsequent differentiation into each mesoderm lineage. (b) Pluripotency was evaluated by staining with OCT4 and SOX2 before the differentiation. hESCs differentiated with CHIR 5 μM were positive for T and TBX6 on day 1.5 of differentiation, but cells did not stain for HOXD11. Scale bars: 200 μm. (c) Immunocytochemistry on day 4 of the differentiation with CHIR (3 to 10 μM). Sustained TBX6 expression was observed when the cells were differentiated with high doses of CHIR (7-10 μM). Scale bar: 100 μm. (d) Real-time PCR for MIXL1 in hESCs from day 0 to 7. hESCs were differentiated with CHIR 8 μM for 4 days, and activin 10 ng/ml for 3 days. MIXL1, another marker for primitive streak also showed sustained expression at least until day 4 of the differentiation. Expression returned to very low levels by day 7. n=2. (e) Staining with WT1 and HOXD11 on day 7 of the differentiation. hESCs were differentiated with CHIR 8 μM for 4 days and subsequently with the basic medium (ARPMI) for 3 days. Cells expressed WT1, but not HOXD11. Scale bar 100 μm.

Supplementary Figure 3 Protocol adjustment in hiPSCs

(a) Comparison of CHIR dose in T expression on day 4. A slightly higher dose of CHIR was required for sustained T expression in hiPSCs on day 4. (b) Immunocytochemistry for T, TBX6 or FOXF1 on day 4 of differentiation. hESCs were differentiated with CHIR 8 μM, and hiPSCs were differentiated with CHIR 10 μM. Notably, FOXF1 was negative in hESCs, but was positive in hiPSCs. (c) The tested protocol and representative immunocytochemistry in hiPSCs. Noggin at >5 ng/ml suppressed FOXF1 expression on day 4. To induce WT1 expression on day 7, Noggin 5 ng/ml was optimal. (d) The differentiation protocol and staining for FOXF1 on day 4, and for WT1 on day 7 in hESCs. Either additional Noggin or BMP4 significantly suppressed WT1 expression on day 7, and BMP4 induced FOXF1 expression on day 4, suggesting endogenous BMP4 signal is optimal in hESCs with CHIR treatment alone. (e) The protocol and staining with WT1 and HOXD11 on day 7 in hiPSCs. CHIR 10 μM + Noggin 5 ng/ml followed by activin 10 ng/ml showed the most efficient differentiation into WT1+HOXD11+ cells in hiPSCs. Scale bars: 100 μm.

Supplementary Figure 4 Spontaneous differentiation of SIX2+ cells into nephrons and growth factor screening in 3D culture

(a) Staining with PAX8 and LHX1 in hESCs on day 10 of the differentiation. hESCs were differentiated with CHIR 8 μM for 4 days, activn 10 ng/ml for 3 days, and FGF9 10 ng/ml for 3 days. Sporadic expression of LHX1 was observed in PAX8+ cells. Scale bar: 50 μm. (b) Brighfield imaging for hESCs on day 10 and 21 of the differentiation. FGF9 was withdrawn on day 10, and cells were cultured in the basic medium by day 21. Scale bars: 50 μm. (c) Brightfield and immunocytochemistry for LTL and NPHS1 in hESCs on day 28 of the differentiation. Scale bar: 50 μm. (d) The protocol for growth factor screening. Cells were replated to 3D culture on day 10, and the listed growth factors and small molecules were tested. (e) The number of LTL+ tubules in the organoids. HGF showed a tendency to increase LTL+ tubules. n=2. (f) Brightfield imaging of 3D co-culture.with an ureteric bud cell sphere. Scale bar: 100 μm. (g) Whole-mountstaining for LTL in the organoids on day 16. Scale bar: 100 μm. (h) Immunohistochemistry of the organoids on day 16. Scale bars: 50 μm. LTL: lotus tetragonolobus lectin. NPHS1: Nephrin.

Supplementary Figure 5 Screening for growth factors and small molecules to induce renal vesicles

(a, b) The tested protocols for renal vesicle induction. (c) Immunocytochemistry for SIX2 and LHX1 in structures on day 14 of differentiation. Transient treatment with CHIR 3 μM from day 9 to 11, in combination with FGF9 10 ng/ml from day 7 to 14, increased the number of LHX1+ cells and suppressed SIX2 expression, suggesting mesenchymal epithelial transition. Scale bars: 50 μm. REGM: renal cell growth medium (Lonza, #CC-3190).

Supplementary Figure 6 Kidney development analysis

(a) DAPT was used to suppress Notch signaling from day 14 to 21. (b) CDH1, PODXL and LTL expression on day 21 in cells derived from hESCs in 2D culture. Scale bars: 50 μm. (c) Percentage of LTL+ nephron structures in control and DAPT-treated on day 21. The nephron number was counted as CDH1+ tubules from 10 fields (x20 magnification) of each sample (n=2). CDH1: Cadherin-1 (E-cadherin). PODXL: Podocalyxin-like (Podocalyxin).

Supplementary Figure 7 Nephrotoxic assay

(A) The protocol for the nephrotoxic assay. Gentamicin 5 mg/ml was added from day 21 to 23. (B) Whole-mount staining on day 23 for CDH1, KIM-1 and LTL in kidney organoids derived from hESCs. Scale bars: 50 μm. CDH1: cadherin-1 (E-cadherin). PODXL: podocalyxin-like (Podocalyxin). LTL: lotus tetragonolobus lectin. KIM-1: kidney injury molecule-1.

Supplementary information

Supplementary Figures and Tables

Supplementary Figures 1–7 and Supplementary Tables 1–3 (PDF 5321 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Morizane, R., Lam, A., Freedman, B. et al. Nephron organoids derived from human pluripotent stem cells model kidney development and injury. Nat Biotechnol 33, 1193–1200 (2015). https://doi.org/10.1038/nbt.3392

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3392

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