DOI QR코드

DOI QR Code

Applications of kidney organoids derived from human pluripotent stem cells

  • Kim, Yong Kyun (Cell Death Disease Research Center, The Catholic University of Korea) ;
  • Nam, Sun Ah (Cell Death Disease Research Center, The Catholic University of Korea) ;
  • Yang, Chul Woo (Department of Internal Medicine, College of Medicine, The Catholic University of Korea)
  • Received : 2018.06.11
  • Accepted : 2018.06.18
  • Published : 2018.07.01

Abstract

The establishment of protocols to differentiate kidney organoids from human pluripotent stem cells provides potential applications of kidney organoids in regenerative medicine. Modeling of renal diseases, drug screening, nephrotoxicity testing of compounds, and regenerative therapy are attractive applications. Although much progress still remains to be made in the development of kidney organoids, recent advances in clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR-associated system 9 (Cas9) genome editing and three-dimensional bioprinting technologies have contributed to the application of kidney organoids in clinical fields. In this section, we review recent advances in the applications of kidney organoids to kidney disease modelling, drug screening, nephrotoxicity testing, and regenerative therapy.

Keywords

Acknowledgement

Supported by : National Research Foundation of Korea (NRF)

References

  1. Morizane R, Lam AQ, Freedman BS, Kishi S, Valerius MT, Bonventre JV. Nephron organoids derived from human pluripotent stem cells model kidney development and injury. Nat Biotechnol 2015;33:1193-1200. https://doi.org/10.1038/nbt.3392
  2. Taguchi A, Kaku Y, Ohmori T, et al. Redefining the in vivo origin of metanephric nephron progenitors enables generation of complex kidney structures from pluripotent stem cells. Cell Stem Cell 2014;14:53-67. https://doi.org/10.1016/j.stem.2013.11.010
  3. Takasato M, Er PX, Becroft M, et al. Directing human embryonic stem cell differentiation towards a renal lineage generates a self-organizing kidney. Nat Cell Biol 2014;16:118-126. https://doi.org/10.1038/ncb2894
  4. Takasato M, Er PX, Chiu HS, et al. Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature 2015;526:564-568. https://doi.org/10.1038/nature15695
  5. Kim YK, Refaeli I, Brooks CR, et al. Gene-edited human kidney organoids reveal mechanisms of disease in podocyte development. Stem Cells 2017;35:2366-2378. https://doi.org/10.1002/stem.2707
  6. Peters DJ, Breuning MH. Autosomal dominant polycystic kidney disease: modification of disease progression. Lancet 2001;358:1439-1444. https://doi.org/10.1016/S0140-6736(01)06531-X
  7. Willey CJ, Blais JD, Hall AK, Krasa HB, Makin AJ, Czerwiec FS. Prevalence of autosomal dominant polycystic kidney disease in the European Union. Nephrol Dial Transplant 2017;32:1356-1363.
  8. Cruz NM, Song X, Czerniecki SM, et al. Organoid cystogenesis reveals a critical role of microenvironment in human polycystic kidney disease. Nat Mater 2017;16:1112-1119. https://doi.org/10.1038/nmat4994
  9. Freedman BS, Brooks CR, Lam AQ, et al. Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nat Commun 2015;6:8715. https://doi.org/10.1038/ncomms9715
  10. Freedman BS. Modeling kidney disease with iPS cells. Biomark Insights 2015;10(Suppl 1):153-169.
  11. Romagnani P. Organoids: modelling polycystic kidney disease. Nat Mater 2017;16:1058-1059. https://doi.org/10.1038/nmat5013
  12. Happe H, Peters DJ. Translational research in ADPKD: lessons from animal models. Nat Rev Nephrol 2014;10:587-601. https://doi.org/10.1038/nrneph.2014.137
  13. Carone FA, Nakamura S, Bacallao R, Nelson WJ, Khokha M, Kanwar YS. Impaired tubulogenesis of cyst-derived cells from autosomal dominant polycystic kidneys. Kidney Int 1995;47:861-868. https://doi.org/10.1038/ki.1995.129
  14. Cruz NM, Freedman BS. CRISPR gene editing in the kidney. Am J Kidney Dis 2018;71:874-883. https://doi.org/10.1053/j.ajkd.2018.02.347
  15. De Vriese AS, Sethi S, Nath KA, Glassock RJ, Fervenza FC. Differentiating primary, genetic, and secondary FSGS in adults: a clinicopathologic approach. J Am Soc Nephrol 2018;29:759-774.
  16. Devarajan P, Spitzer A. Towards a biological characterization of focal segmental glomerulosclerosis. Am J Kidney Dis 2002;39:625-636. https://doi.org/10.1053/ajkd.2002.31420
  17. Kang HG, Lee M, Lee KB, et al. Loss of podocalyxin causes a novel syndromic type of congenital nephrotic syndrome. Exp Mol Med 2017;49:e414. https://doi.org/10.1038/emm.2017.227
  18. Devuyst O, Knoers NV, Remuzzi G, Schaefer F; Board of the Working Group for Inherited Kidney Diseases of the European Renal Association and European Dialysis and Transplant Association. Rare inherited kidney diseases: challenges, opportunities, and perspectives. Lancet 2014;383:1844-1859. https://doi.org/10.1016/S0140-6736(14)60659-0
  19. Tiong HY, Huang P, Xiong S, Li Y, Vathsala A, Zink D. Drug-induced nephrotoxicity: clinical impact and preclinical in vitro models. Mol Pharm 2014;11:1933-1948. https://doi.org/10.1021/mp400720w
  20. Wilmer MJ, Ng CP, Lanz HL, Vulto P, Suter-Dick L, Masereeuw R. Kidney-on-a-chip technology for drug-induced nephrotoxicity screening. Trends Biotechnol 2016;34:156-170. https://doi.org/10.1016/j.tibtech.2015.11.001
  21. Anzai N, Jutabha P, Kanai Y, Endou H. Integrated physiology of proximal tubular organic anion transport. Curr Opin Nephrol Hypertens 2005;14:472-479. https://doi.org/10.1097/01.mnh.0000170751.56527.7e
  22. Morrissey KM, Stocker SL, Wittwer MB, Xu L, Giacomini KM. Renal transporters in drug development. Annu Rev Pharmacol Toxicol 2013;53:503-529. https://doi.org/10.1146/annurev-pharmtox-011112-140317
  23. Li Y, Oo ZY, Chang SY, et al. An in vitro method for the prediction of renal proximal tubular toxicity in humans. Toxicol Res 2013;2:352-365. https://doi.org/10.1039/c3tx50042j
  24. Su R, Li Y, Zink D, Loo LH. Supervised prediction of drug-induced nephrotoxicity based on interleukin-6 and -8 expression levels. BMC Bioinformatics 2014;15 Suppl 16:S16.
  25. Jenkinson SE, Chung GW, van Loon E, Bakar NS, Dalzell AM, Brown CD. The limitations of renal epithelial cell line HK-2 as a model of drug transporter expression and function in the proximal tubule. Pflugers Arch 2012;464:601-611. https://doi.org/10.1007/s00424-012-1163-2
  26. Mutsaers HA, Wilmer MJ, van den Heuvel LP, Hoenderop JG, Masereeuw R. Basolateral transport of the uraemic toxin p-Cresyl sulfate: role for organic anion transporters? Nephrol Dial Transplant 2011;26:4149. https://doi.org/10.1093/ndt/gfr562
  27. Slusser A, Bathula CS, Sens DA, et al. Cadherin expression, vectorial active transport, and metallothionein isoform 3 mediated EMT/MET responses in cultured primary and immortalized human proximal tubule cells. PLoS One 2015;10:e0120132. https://doi.org/10.1371/journal.pone.0120132
  28. Brown CD, Sayer R, Windass AS, et al. Characterisation of human tubular cell monolayers as a model of proximal tubular xenobiotic handling. Toxicol Appl Pharmacol 2008;233:428-438. https://doi.org/10.1016/j.taap.2008.09.018
  29. Lash LH, Putt DA, Cai H. Drug metabolism enzyme expression and activity in primary cultures of human proximal tubular cells. Toxicology 2008;244:56-65. https://doi.org/10.1016/j.tox.2007.10.022
  30. Van der Hauwaert C, Savary G, Gnemmi V, et al. Isolation and characterization of a primary proximal tubular epithelial cell model from human kidney by CD10/CD13 double labeling. PLoS One 2013;8:e66750. https://doi.org/10.1371/journal.pone.0066750
  31. Li Y, Kandasamy K, Chuah JK, et al. Identification of nephrotoxic compounds with embryonic stem-cellderived human renal proximal tubular-like cells. Mol Pharm 2014;11:1982-1990. https://doi.org/10.1021/mp400637s
  32. Narayanan K, Schumacher KM, Tasnim F, et al. Human embryonic stem cells differentiate into functional renal proximal tubular-like cells. Kidney Int 2013;83:593-603. https://doi.org/10.1038/ki.2012.442
  33. Kandasamy K, Chuah JK, Su R, et al. Prediction of drug-induced nephrotoxicity and injury mechanisms with human induced pluripotent stem cell-derived cells and machine learning methods. Sci Rep 2015;5:12337. https://doi.org/10.1038/srep12337
  34. Cukierman E, Pankov R, Stevens DR, Yamada KM. Taking cell-matrix adhesions to the third dimension. Science 2001;294:1708-1712. https://doi.org/10.1126/science.1064829
  35. Cukierman E, Pankov R, Yamada KM. Cell interactions with three-dimensional matrices. Curr Opin Cell Biol 2002;14:633-639. https://doi.org/10.1016/S0955-0674(02)00364-2
  36. Geiger B, Bershadsky A, Pankov R, Yamada KM. Transmembrane crosstalk between the extracellular matrix: cytoskeleton crosstalk. Nat Rev Mol Cell Biol 2001;2:793-805. https://doi.org/10.1038/35099066
  37. Astashkina AI, Mann BK, Prestwich GD, Grainger DW. A 3-D organoid kidney culture model engineered for high-throughput nephrotoxicity assays. Biomaterials 2012;33:4700-4711. https://doi.org/10.1016/j.biomaterials.2012.02.063
  38. Su R, Xiong S, Zink D, Loo LH. High-throughput imaging-based nephrotoxicity prediction for xenobiotics with diverse chemical structures. Arch Toxicol 2016;90:2793-2808. https://doi.org/10.1007/s00204-015-1638-y
  39. Ramm S, Adler M, Vaidya VS. A high-throughput screening assay to identify kidney toxic compounds. Curr Protoc Toxicol 2016;69:9.
  40. Czerniecki SM, Cruz NM, Harder JL, et al. High-throughput screening enhances kidney organoid differentiation from human pluripotent stem cells and enables automated multidimensional phenotyping. Cell Stem Cell 2018;22:929-940. https://doi.org/10.1016/j.stem.2018.04.022
  41. Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nat Biotechnol 2014;32:773-785. https://doi.org/10.1038/nbt.2958
  42. Homan KA, Kolesky DB, Skylar-Scott MA, et al. Bioprinting of 3D convoluted renal proximal tubules on perfusable chips. Sci Rep 2016;6:34845. https://doi.org/10.1038/srep34845
  43. Osaki T, Sivathanu V, Kamm RD. Vascularized microfluidic organ-chips for drug screening, disease models and tissue engineering. Curr Opin Biotechnol 2018;52:116-123. https://doi.org/10.1016/j.copbio.2018.03.011
  44. King S, Creasey O, Presnell S, Nguyen D. Design and characterization of a multicellular, three-dimensional (3D) tissue model of the human kidney proximal tubule FASEB J 2015;29(1 Suppl):LB426.
  45. Atala A, Bauer SB, Soker S, Yoo JJ, Retik AB. Tissue-engineered autologous bladders for patients needing cystoplasty. Lancet 2006;367:1241-1246. https://doi.org/10.1016/S0140-6736(06)68438-9
  46. Chang JW, Park SA, Park JK, et al. Tissue-engineered tracheal reconstruction using three-dimensionally printed artificial tracheal graft: preliminary report. Artif Organs 2014;38:E95-E105. https://doi.org/10.1111/aor.12310
  47. Chuah JKC, Zink D. Stem cell-derived kidney cells and organoids: recent breakthroughs and emerging applications. Biotechnol Adv 2017;35:150-167. https://doi.org/10.1016/j.biotechadv.2016.12.001
  48. Jha V, Garcia-Garcia G, Iseki K, et al. Chronic kidney disease: global dimension and perspectives. Lancet 2013; 382:260-272. https://doi.org/10.1016/S0140-6736(13)60687-X
  49. Jin DC, Yun SR, Lee SW, et al. Current characteristics of dialysis therapy in Korea: 2016 registry data focusing on diabetic patients. Kidney Res Clin Pract 2018;37:20-29. https://doi.org/10.23876/j.krcp.2018.37.1.20
  50. Jin DC, Yun SR, Lee SW, et al. Lessons from 30 years' data of Korean end-stage renal disease registry, 1985-2015. Kidney Res Clin Pract 2015;34:132-139. https://doi.org/10.1016/j.krcp.2015.08.004
  51. Kiuchi MG, Mion D Jr. Chronic kidney disease and risk factors responsible for sudden cardiac death: a whiff of hope? Kidney Res Clin Pract 2016;35:3-9. https://doi.org/10.1016/j.krcp.2015.11.003
  52. Jeong HY, Cho HJ, Kim SH, et al. Association of serum uric acid level with coronary artery stenosis severity in Korean end-stage renal disease patients. Kidney Res Clin Pract 2017;36:282-289. https://doi.org/10.23876/j.krcp.2017.36.3.282
  53. Bae EH, Kim HY, Kang YU, Kim CS, Ma SK, Kim SW. Risk factors for in-hospital mortality in patients starting hemodialysis. Kidney Res Clin Pract 2015;34:154-159. https://doi.org/10.1016/j.krcp.2015.07.005
  54. Imberti B, Tomasoni S, Ciampi O, et al. Renal progenitors derived from human iPSCs engraft and restore function in a mouse model of acute kidney injury. Sci Rep 2015;5:8826. https://doi.org/10.1038/srep08826
  55. Sharmin S, Taguchi A, Kaku Y, et al. Human induced pluripotent stem cell-derived podocytes mature into vascularized glomeruli upon experimental transplantation. J Am Soc Nephrol 2016;27:1778-1791. https://doi.org/10.1681/ASN.2015010096
  56. Toyohara T, Mae S, Sueta S, et al. Cell therapy using human induced pluripotent stem cell-derived renal progenitors ameliorates acute kidney injury in mice. Stem Cells Transl Med 2015;4:980-992. https://doi.org/10.5966/sctm.2014-0219
  57. van den Berg CW, Ritsma L, Avramut MC, et al. Renal subcapsular transplantation of PSC-derived kidney organoids induces neo-vasculogenesis and significant glomerular and tubular maturation in vivo. Stem Cell Reports 2018;10:751-765. https://doi.org/10.1016/j.stemcr.2018.01.041
  58. Gonfiotti A, Jaus MO, Barale D, et al. The first tissue-engineered airway transplantation: 5-year follow-up results. Lancet 2014;383:238-244. https://doi.org/10.1016/S0140-6736(13)62033-4
  59. Macchiarini P, Jungebluth P, Go T, Asnaghi MA, et al. Clinical transplantation of a tissue-engineered airway. Lancet 2008;372:2023-2030. https://doi.org/10.1016/S0140-6736(08)61598-6
  60. Song JJ, Guyette JP, Gilpin SE, Gonzalez G, Vacanti JP, Ott HC. Regeneration and experimental orthotopic transplantation of a bioengineered kidney. Nat Med 2013;19:646-651. https://doi.org/10.1038/nm.3154
  61. Bonandrini B, Figliuzzi M, Papadimou E, et al. Recellularization of well-preserved acellular kidney scaffold using embryonic stem cells. Tissue Eng Part A 2014;20:1486-1498. https://doi.org/10.1089/ten.tea.2013.0269
  62. Guan Y, Liu S, Sun C, et al. The effective bioengineering method of implantation decellularized renal extracellular matrix scaffolds. Oncotarget 2015;6:36126-36138.
  63. Caralt M, Uzarski JS, Iacob S, et al. Optimization and critical evaluation of decellularization strategies to develop renal extracellular matrix scaffolds as biological templates for organ engineering and transplantation. Am J Transplant 2015;15:64-75. https://doi.org/10.1111/ajt.12999
  64. Hachisuka S, Sato Y, Yoshiike M, Nakazawa R, Sasaki H, Chikaraishi T. Enhanced recellularization of renal extracellular matrix scaffold under negative pressure. Integr Mol Med 2015;2:394-99.
  65. Du C, Narayanan K, Leong MF, et al. Functional kidney bioengineering with pluripotent stem-cell-derived renal progenitor cells and decellularized kidney scaffolds. Adv Healthc Mater 2016;5:2080-2091. https://doi.org/10.1002/adhm.201600120
  66. Sampaio FJ, Pereira-Sampaio MA, Favorito LA. The pig kidney as an endourologic model: anatomic contribution. J Endourol 1998;12:45-50. https://doi.org/10.1089/end.1998.12.45
  67. Wilm B, Tamburrini R, Orlando G, Murray P. Autologous cells for kidney bioengineering. Curr Transplant Rep 2016;3:207-220. https://doi.org/10.1007/s40472-016-0107-8
  68. Morizane R, Bonventre JV. Kidney organoids: a translational journey. Trends Mol Med 2017;23:246-263. https://doi.org/10.1016/j.molmed.2017.01.001

Cited by

  1. Mechanobiology of cells and cell systems, such as organoids vol.11, pp.5, 2018, https://doi.org/10.1007/s12551-019-00590-7
  2. Graft immaturity and safety concerns in transplanted human kidney organoids vol.51, pp.11, 2019, https://doi.org/10.1038/s12276-019-0336-x
  3. Hydrogel-integrated Microfluidic Systems for Advanced Stem Cell Engineering vol.13, pp.4, 2018, https://doi.org/10.1007/s13206-019-3402-5
  4. A glomerulus-on-a-chip to recapitulate the human glomerular filtration barrier vol.10, pp.1, 2019, https://doi.org/10.1038/s41467-019-11577-z
  5. Mouse Embryonic Stem Cell-Derived Ureteric Bud Progenitors Induce Nephrogenesis vol.9, pp.2, 2018, https://doi.org/10.3390/cells9020329
  6. Most Influential Physicochemical and In Vitro Assay Descriptors for Hepatotoxicity and Nephrotoxicity Prediction vol.33, pp.7, 2020, https://doi.org/10.1021/acs.chemrestox.0c00040
  7. Advances in development and application of human organoids vol.11, pp.6, 2021, https://doi.org/10.1007/s13205-021-02815-7
  8. Human kidney organoids reveal the role of glutathione in Fabry disease vol.53, pp.10, 2018, https://doi.org/10.1038/s12276-021-00683-y
  9. Organoids: A new approach in toxicity testing of nanotherapeutics vol.42, pp.1, 2018, https://doi.org/10.1002/jat.4206
  10. Drug-Induced Nephrotoxicity Assessment in 3D Cellular Models vol.13, pp.1, 2018, https://doi.org/10.3390/mi13010003