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.

Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells

This article has been updated

Abstract

Transplantation of pancreatic progenitors or insulin-secreting cells derived from human embryonic stem cells (hESCs) has been proposed as a therapy for diabetes. We describe a seven-stage protocol that efficiently converts hESCs into insulin-producing cells. Stage (S) 7 cells expressed key markers of mature pancreatic beta cells, including MAFA, and displayed glucose-stimulated insulin secretion similar to that of human islets during static incubations in vitro. Additional characterization using single-cell imaging and dynamic glucose stimulation assays revealed similarities but also notable differences between S7 insulin-secreting cells and primary human beta cells. Nevertheless, S7 cells rapidly reversed diabetes in mice within 40 days, roughly four times faster than pancreatic progenitors. Therefore, although S7 cells are not fully equivalent to mature beta cells, their capacity for glucose-responsive insulin secretion and rapid reversal of diabetes in vivo makes them a promising alternative to pancreatic progenitor cells or cadaveric islets for the treatment of diabetes.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Overview of differentiation protocol and characterization of S4–S6.
Figure 2: S6 insulin+ cells co-express key transcription factors, such as NKX6.1, unlike S4 pancreatic progenitor cells.
Figure 3: S6 insulin+ cells develop faster in vivo than S4 pancreatic progenitor cells.
Figure 4: Optimization of S7 differentiation protocol for induction of MAFA expression.
Figure 5: Stage 7 cells have a similar gene expression profile to human islets, and insulin+ cells co-express key beta-cell maturation markers, including MAFA.
Figure 6: Stage 7 cells have certain functional similarities to human beta cells, but cannot be considered mature human beta cells.
Figure 7: S7 cells rapidly reverse diabetes and develop glucose-responsive insulin secretion in vivo.

Change history

  • 16 September 2014

    Erratum: In the version of this article initially published online, there were several errors in the PDF and/or the HTML versions. PDF and HTML versions: In the abstract, the last sentence should have read “in vivo” rather than “in vitro.” In the Methods, under the heading “Differentiation of S4 cells in suspension aggregate cultures,” the units of the cell clump size should have been micrometers rather than millimeters, as follows: “On the last day of culture, S4 cells were treated with 5 mg/ml dispase (Corning, Cat# 354235) for 5 min at 37 °C, followed by gentle pipetting to break into cell clumps (100–200 μm).” In the Methods, under the heading “S4: pancreatic endoderm, PDX1+/NKX6.1+ cells (3 d),” milliliter should have been microliter, as follows: “The resulting cell pellet was resuspended as single cells at a density of ~0.5 × 105 cells/μl on filter inserts (BD, Cat#35-3493 or Corning Cat#3420); 5–10 μl per spot for a total of 0.25–0.5 × 106 cells/spot) at an air-liquid interface.” HTML version: In the legends to Figures 2, 3, 4 and 7, the units of the scale bars should have been micrometers rather than millimeters. The errors have been corrected for the print, PDF and HTML versions of this article.

References

  1. Barton, F.B. et al. Improvement in outcomes of clinical islet transplantation: 1999–2010. Diabetes Care 35, 1436–1445 (2012).

    Article  CAS  Google Scholar 

  2. Warnock, G.L. et al. A multi-year analysis of islet transplantation compared with intensive medical therapy on progression of complications in type 1 diabetes. Transplantation 86, 1762–1766 (2008).

    Article  Google Scholar 

  3. Bruin, J.E. et al. Characterization of polyhormonal insulin-producing cells derived in vitro from human embryonic stem cells. Stem Cell Res. 12, 194–208 (2014).

    Article  CAS  Google Scholar 

  4. Bruin, J.E. et al. Maturation and function of human embryonic stem cell-derived pancreatic progenitors in macroencapsulation devices following transplant into mice. Diabetologia 56, 1987–1998 (2013).

    Article  Google Scholar 

  5. Rezania, A. et al. Maturation of human embryonic stem cell-derived pancreatic progenitors into functional islets capable of treating pre-existing diabetes in mice. Diabetes 61, 2016–2029 (2012).

    Article  CAS  Google Scholar 

  6. Rezania, A. et al. Enrichment of human embryonic stem cell-derived NKX6.1-expressing pancreatic progenitor cells accelerates the maturation of insulin-secreting cells in vivo. Stem Cells 31, 2432–2442 (2013).

    Article  CAS  Google Scholar 

  7. Rezania, A. et al. Production of functional glucagon-secreting alpha-cells from human embryonic stem cells. Diabetes 60, 239–247 (2011).

    Article  CAS  Google Scholar 

  8. Chen, S. et al. A small molecule that directs differentiation of human ESCs into the pancreatic lineage. Nat. Chem. Biol. 5, 258–265 (2009).

    Article  CAS  Google Scholar 

  9. Xu, X., Browning, V.L. & Odorico, J.S. Activin, BMP and FGF pathways cooperate to promote endoderm and pancreatic lineage cell differentiation from human embryonic stem cells. Mech. Dev. 128, 412–427 (2011).

    Article  Google Scholar 

  10. Shim, J.H. et al. Directed differentiation of human embryonic stem cells towards a pancreatic cell fate. Diabetologia 50, 1228–1238 (2007).

    Article  CAS  Google Scholar 

  11. Cho, C.H. et al. Inhibition of activin/nodal signalling is necessary for pancreatic differentiation of human pluripotent stem cells. Diabetologia 55, 3284–3295 (2012).

    Article  CAS  Google Scholar 

  12. D'Amour, K.A. et al. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat. Biotechnol. 24, 1392–1401 (2006).

    Article  CAS  Google Scholar 

  13. Kroon, E. et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat. Biotechnol. 26, 443–452 (2008).

    Article  CAS  Google Scholar 

  14. Nostro, M.C. et al. Stage-specific signaling through TGFbeta family members and WNT regulates patterning and pancreatic specification of human pluripotent stem cells. Development 138, 861–871 (2011).

    Article  CAS  Google Scholar 

  15. Kelly, O.G. et al. Cell-surface markers for the isolation of pancreatic cell types derived from human embryonic stem cells. Nat. Biotechnol. 29, 750–756 (2011).

    Article  CAS  Google Scholar 

  16. Schiesser, J.V. & Wells, J.M. Generation of beta cells from human pluripotent stem cells: are we there yet? Ann. NY Acad. Sci. 1311, 124–137 (2014).

    Article  CAS  Google Scholar 

  17. Nostro, M.C. & Keller, G. Generation of beta cells from human pluripotent stem cells: Potential for regenerative medicine. Semin. Cell Dev. Biol. 23, 701–710 (2012).

    Article  CAS  Google Scholar 

  18. Halban, P.A., Kahn, S.E., Lernmark, A. & Rhodes, C.J. Gene and cell-replacement therapy in the treatment of type 1 diabetes: how high must the standards be set? Diabetes 50, 2181–2191 (2001).

    Article  CAS  Google Scholar 

  19. Aguayo-Mazzucato, C. & Bonner-Weir, S. Stem cell therapy for type 1 diabetes mellitus. Nat. Rev. Endocrinol. 6, 139–148 (2010).

    Article  Google Scholar 

  20. Wells, J.M. & Melton, D.A. Vertebrate endoderm development. Annu. Rev. Cell Dev. Biol. 15, 393–410 (1999).

    Article  CAS  Google Scholar 

  21. Nelson, S.B., Schaffer, A.E. & Sander, M. The transcription factors Nkx6.1 and Nkx6.2 possess equivalent activities in promoting beta-cell fate specification in Pdx1+ pancreatic progenitor cells. Development 134, 2491–2500 (2007).

    Article  CAS  Google Scholar 

  22. Sander, M. et al. Homeobox gene Nkx6.1 lies downstream of Nkx2.2 in the major pathway of beta-cell formation in the pancreas. Development 127, 5533–5540 (2000).

    CAS  PubMed  Google Scholar 

  23. Rukstalis, J.M. & Habener, J.F. Neurogenin3: a master regulator of pancreatic islet differentiation and regeneration. Islets 1, 177–184 (2009).

    Article  Google Scholar 

  24. Matsuoka, T.A. et al. The MafA transcription factor appears to be responsible for tissue-specific expression of insulin. Proc. Natl. Acad. Sci. USA 101, 2930–2933 (2004).

    Article  CAS  Google Scholar 

  25. Wang, H., Brun, T., Kataoka, K., Sharma, A.J. & Wollheim, C.B. MAFA controls genes implicated in insulin biosynthesis and secretion. Diabetologia 50, 348–358 (2007).

    Article  CAS  Google Scholar 

  26. Zhang, C. et al. MafA is a key regulator of glucose-stimulated insulin secretion. Mol. Cell. Biol. 25, 4969–4976 (2005).

    Article  CAS  Google Scholar 

  27. Hang, Y. et al. The MafA transcription factor becomes essential to islet beta-cells soon after birth. Diabetes 63, 1994–2005 (2014).

    Article  CAS  Google Scholar 

  28. Choi, K.M. et al. Effect of ascorbic acid on bone marrow-derived mesenchymal stem cell proliferation and differentiation. J. Biosci. Bioeng. 105, 586–594 (2008).

    Article  CAS  Google Scholar 

  29. Johansson, K.A. et al. Temporal control of neurogenin3 activity in pancreas progenitors reveals competence windows for the generation of different endocrine cell types. Dev. Cell 12, 457–465 (2007).

    Article  CAS  Google Scholar 

  30. Schulz, T.C. et al. A scalable system for production of functional pancreatic progenitors from human embryonic stem cells. PLoS ONE 7, e37004 (2012).

    Article  CAS  Google Scholar 

  31. Pruniéras, M., Regnier, M. & Woodley, D. Methods for cultivation of keratinocytes with an air-liquid interface. J. Invest. Dermatol. 81, 28s–33s (1983).

    Article  Google Scholar 

  32. Pezzulo, A.A. et al. The air-liquid interface and use of primary cell cultures are important to recapitulate the transcriptional profile of in vivo airway epithelia. Am. J. Physiol. Lung Cell. Mol. Physiol. 300, L25–L31 (2011).

    Article  CAS  Google Scholar 

  33. Heinis, M. et al. Oxygen tension regulates pancreatic beta-cell differentiation through hypoxia-inducible factor 1alpha. Diabetes 59, 662–669 (2010).

    Article  CAS  Google Scholar 

  34. Cortijo, C., Gouzi, M., Tissir, F. & Grapin-Botton, A. Planar cell polarity controls pancreatic beta cell differentiation and glucose homeostasis. Cell Reports 2, 1593–1606 (2012).

    Article  CAS  Google Scholar 

  35. Shi, Y. & Massagué, J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 113, 685–700 (2003).

    Article  CAS  Google Scholar 

  36. Aguayo-Mazzucato, C. et al. Thyroid hormone promotes postnatal rat pancreatic beta-cell development and glucose-responsive insulin secretion through MAFA. Diabetes 62, 1569–1580 (2013).

    Article  CAS  Google Scholar 

  37. Seymour, P.A. et al. SOX9 is required for maintenance of the pancreatic progenitor cell pool. Proc. Natl. Acad. Sci. USA 104, 1865–1870 (2007).

    Article  CAS  Google Scholar 

  38. Murtaugh, L.C., Stanger, B.Z., Kwan, K.M. & Melton, D.A. Notch signaling controls multiple steps of pancreatic differentiation. Proc. Natl. Acad. Sci. USA 100, 14920–14925 (2003).

    Article  CAS  Google Scholar 

  39. Apelqvist, A. et al. Notch signalling controls pancreatic cell differentiation. Nature 400, 877–881 (1999).

    Article  CAS  Google Scholar 

  40. Pisania, A. et al. Quantitative analysis of cell composition and purity of human pancreatic islet preparations. Lab. Invest. 90, 1661–1675 (2010).

    Article  Google Scholar 

  41. Street, C.N. et al. Islet graft assessment in the Edmonton Protocol: implications for predicting long-term clinical outcome. Diabetes 53, 3107–3114 (2004).

    Article  CAS  Google Scholar 

  42. Piper, K. et al. Beta cell differentiation during early human pancreas development. J. Endocrinol. 181, 11–23 (2004).

    Article  CAS  Google Scholar 

  43. Aguayo-Mazzucato, C. et al. Mafa expression enhances glucose-responsive insulin secretion in neonatal rat beta cells. Diabetologia 54, 583–593 (2011).

    Article  CAS  Google Scholar 

  44. Haase, T.N. et al. Growth arrest specific protein (GAS) 6: a role in the regulation of proliferation and functional capacity of the perinatal rat beta cell. Diabetologia 56, 763–773 (2013).

    Article  CAS  Google Scholar 

  45. Harmon, J.S. et al. beta-Cell-specific overexpression of glutathione peroxidase preserves intranuclear MafA and reverses diabetes in db/db mice. Endocrinology 150, 4855–4862 (2009).

    Article  CAS  Google Scholar 

  46. Harmon, J.S., Stein, R. & Robertson, R.P. Oxidative stress-mediated, post-translational loss of MafA protein as a contributing mechanism to loss of insulin gene expression in glucotoxic beta cells. J. Biol. Chem. 280, 11107–11113 (2005).

    Article  CAS  Google Scholar 

  47. Mahadevan, J. et al. Ebselen treatment prevents islet apoptosis, maintains intranuclear Pdx-1 and MafA levels, and preserves beta-cell mass and function in ZDF rats. Diabetes 62, 3582–3588 (2013).

    Article  CAS  Google Scholar 

  48. Merani, S., Toso, C., Emamaullee, J. & Shapiro, A.M. Optimal implantation site for pancreatic islet transplantation. Br. J. Surg. 95, 1449–1461 (2008).

    Article  CAS  Google Scholar 

  49. Kumar, M. & Melton, D. Pancreas specification: a budding question. Curr. Opin. Genet. Dev. 13, 401–407 (2003).

    Article  CAS  Google Scholar 

  50. Jørgensen, M.C. et al. An illustrated review of early pancreas development in the mouse. Endocr. Rev. 28, 685–705 (2007).

    Article  Google Scholar 

  51. Kinkel, M.D. & Prince, V.E. On the diabetic menu: zebrafish as a model for pancreas development and function. BioEssays 31, 139–152 (2009).

    Article  CAS  Google Scholar 

  52. D'Amour, K.A. et al. Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat. Biotechnol. 23, 1534–1541 (2005).

    Article  CAS  Google Scholar 

  53. Basford, C.L. et al. The functional and molecular characterisation of human embryonic stem cell-derived insulin-positive cells compared with adult pancreatic beta cells. Diabetologia 55, 358–371 (2012).

    Article  CAS  Google Scholar 

  54. Luciani, D.S. & Johnson, J.D. Acute effects of insulin on beta-cells from transplantable human islets. Mol. Cell. Endocrinol. 241, 88–98 (2005).

    Article  CAS  Google Scholar 

  55. Misler, S., Barnett, D.W., Gillis, K.D. & Pressel, D.M. Electrophysiology of stimulus-secretion coupling in human beta-cells. Diabetes 41, 1221–1228 (1992).

    Article  CAS  Google Scholar 

  56. Nauck, M.A. et al. Incretin effects of increasing glucose loads in man calculated from venous insulin and C-peptide responses. J. Clin. Endocrinol. Metab. 63, 492–498 (1986).

    Article  CAS  Google Scholar 

  57. Davalli, A.M. et al. A selective decrease in the beta cell mass of human islets transplanted into diabetic nude mice. Transplantation 59, 817–820 (1995).

    Article  CAS  Google Scholar 

  58. Tuch, B.E. Reversal of diabetes by human fetal pancreas. Optimization of requirements in the hyperglycemic nude mouse. Transplantation 51, 557–562 (1991).

    Article  CAS  Google Scholar 

  59. Tuch, B.E. & Monk, R.S. Regulation of blood glucose to human levels by human fetal pancreatic xenografts. Transplantation 51, 1156–1160 (1991).

    Article  CAS  Google Scholar 

  60. Johnson, J.D. et al. Different effects of FK506, rapamycin, and mycophenolate mofetil on glucose-stimulated insulin release and apoptosis in human islets. Cell Transplant. 18, 833–845 (2009).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported in part by funding from the JDRF, the Canadian Institutes of Health Research (CIHR) Regenerative Medicine and Nanomedicine Initiative, and the Stem Cell Network (SCN). J.E.B. was funded by a JDRF postdoctoral fellowship and L'Oreal Canada for Women in Science Research Excellence Fellowship. Y.H.C.Y. was the recipient of an NSERC Post Graduate Scholarship and M.M. was the recipient of a Mitacs Elevate Postdoctoral Fellowship. The CIHR Transplantation Training Program provided funding for J.E.B., N.Q. and M.M. We also thank Stem Cell Technologies for their financial support, T. Webber, S. Karuna and B. Hu for their technical assistance, and G. Warnock and Z. Ao from the Irving K. Barber Human Islet Isolation Laboratory (Vancouver, BC) for providing human islets.

Author information

Authors and Affiliations

Authors

Contributions

A. Rezania, J.E.B., J.D.J. and T.J.K. designed experiments, analyzed and interpreted results. J.E.B., P.A., A. Rubin, I.B., A.A., S.O., N.Q., M.M., T.A. and Y.H.C.Y. performed experiments. A. Rezania and T.J.K. conceived the project and supervised its participants. A. Rezania, J.E.B., J.D.J. and T.J.K. wrote the manuscript. A. Rezania, J.E.B., A.A., J.D.J., T.J.K., N.Q., M.M., T.A. and Y.H.C.Y. contributed to manuscript revisions.

Corresponding authors

Correspondence to Alireza Rezania or Timothy J Kieffer.

Ethics declarations

Competing interests

A. Rezania, P.A., A. Rubin and I.B. are employees of Janssen R&D, LLC; T.J.K. received financial support from Janssen R&D, LLC, for the research described in this article.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–26 and Supplementary Tables 1–8 (PDF 16303 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rezania, A., Bruin, J., Arora, P. et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol 32, 1121–1133 (2014). https://doi.org/10.1038/nbt.3033

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3033

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