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.

  • Review Article
  • Published:

The use of stem cells for pancreatic regeneration in diabetes mellitus

Abstract

The endocrine pancreas represents an interesting arena for regenerative medicine and cell therapeutics. One of the major pancreatic diseases, diabetes mellitus is a metabolic disorder caused by having an insufficient number of insulin-producing β cells. Replenishment of β cells by cell transplantation can restore normal metabolic control. The shortage in donor pancreata has meant that the demand for transplantable β cells has outstripped the supply, which could be met by using alternative sources of stem cells. This situation has opened up new areas of research, such as cellular reprogramming and in vivo β-cell regeneration. Pluripotent stem cells seem to be the best option for clinical applications of β-cell regeneration in the near future, as these cells have been demonstrated to represent an unlimited source of functional β cells. Although compelling evidence shows that the adult pancreas retains regenerative capacity, it remains unclear whether this organ contains stem cells. Alternatively, specialized cell types within or outside the pancreas retain plasticity in proliferation and differentiation. Cellular reprogramming or transdifferentiation of exocrine cells or other types of endocrine cells in the pancreas could provide a long-term solution.

Key Points

  • Pancreatic progenitors can be derived from human embryonic stem cells and have the capacity to generate functional β cells after transplantation

  • Whether mesenchymal stem cells and other types of adult stem cells can generate functional β cells remains unclear

  • Some types of differentiated cells within the adult pancreas retain sufficient plasticity to transdifferentiate into β cells; the most promising examples are α cells and acinar exocrine cells

  • The available data on pancreatic β-cell regeneration and transdifferentiation have mainly been obtained in mouse models and still need to be reproduced in humans

  • The future success of transplantation of β cells derived from embryonic stem cells or regeneration of endogenous β cells depends on effective prevention of autoimmune and/or alloimmune rejection

  • The most promising strategy at the moment is transplantation of pancreatic progenitors (or β cells) derived from embryonic stem cells, as these are widely available and standard protocols already exist

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: Current strategies for generating pancreatic (endocrine) progenitors from human pluripotent stem cells.
Figure 2: Candidate progenitor cells, from which pancreatic β cells could be derived.

Similar content being viewed by others

Zixuan Zhao, Xinyi Chen, … Hanry Yu

References

  1. Weir, G. C. & Bonner-Weir, S. Islet β-cell mass in diabetes and how it relates to function, birth, and death. Ann. NY Acad. Sci. 1281, 92–105 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Butler, A. E. et al. Modestly increased β-cell apoptosis but no increased β-cell replication in recent-onset type 1 diabetic patients who died of diabetic ketoacidosis. Diabetologia 50, 2323–2331 (2007).

    Article  CAS  PubMed  Google Scholar 

  3. Gepts, W. Pathologic anatomy of the pancreas in juvenile diabetes mellitus. Diabetes 14, 619–633 (1965).

    Article  CAS  PubMed  Google Scholar 

  4. Pipeleers, D. & Ling, Z. Pancreatic β cells in insulin-dependent diabetes. Diabetes Metab. Rev. 8, 209–227 (1992).

    Article  CAS  PubMed  Google Scholar 

  5. American diabetes association. Diagnosis and classification of diabetes mellitus. Diabetes Care 36 (Suppl. 1), S67–S74 (2013).

  6. Butler, A. E. et al. β-cell deficit and increased β-cell apoptosis in humans with type 2 diabetes. Diabetes 52, 102–110 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Rahier, J., Guiot, Y., Goebbels, R. M., Sempoux, C. & Henquin, J. C. Pancreatic β-cell mass in European subjects with type 2 diabetes. Diabetes Obes. Metab. 10 (Suppl. 4), 32–42 (2008).

    Article  PubMed  Google Scholar 

  8. [No authors listed] The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The diabetes control and complications trial research group. N. Engl. J. Med. 329, 977–986 (1993).

  9. Keymeulen, B. et al. Implantation of standardized β-cell grafts in a liver segment of IDDM patients: graft and recipients characteristics in two cases of insulin-independence under maintenance immunosuppression for prior kidney graft. Diabetologia 41, 452–459 (1998).

    Article  CAS  PubMed  Google Scholar 

  10. Ryan, E. A. et al. Clinical outcomes and insulin secretion after islet transplantation with the Edmonton protocol. Diabetes 50, 710–719 (2001).

    Article  CAS  PubMed  Google Scholar 

  11. Shapiro, A. M. et al. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N. Engl. J. Med. 343, 230–238 (2000).

    Article  CAS  PubMed  Google Scholar 

  12. Keymeulen, B. et al. Correlation between β-cell mass and glycemic control in type 1 diabetic recipients of islet cell graft. Proc. Natl Acad. Sci. USA 103, 17444–17449 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ryan, E. A. et al. Five-year follow-up after clinical islet transplantation. Diabetes 54, 2060–2069 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Jacobs-Tulleneers-Thevissen, D. et al. Human islet cell implants in a nude rat model of diabetes survive better in omentum than in liver with a positive influence of β-cell number and purity. Diabetologia 53, 1690–1699 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. van der Windt, D. J., Echeverri, G. J., Ijzermans, J. N. & Cooper, D. K. The choice of anatomical site for islet transplantation. Cell Transplant. 17, 1005–1014 (2008).

    Article  PubMed  Google Scholar 

  16. Parsons, R. F. et al. Murine islet allograft tolerance upon blockade of the B-lymphocyte stimulator, BLyS/BAFF. Transplantation 93, 676–685 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Takiishi, T. et al. Reversal of autoimmune diabetes by restoration of antigen-specific tolerance using genetically modified Lactococcus lactis in mice. J. Clin. Invest. 122, 1717–1725 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. You, S. et al. Induction of allograft tolerance by monoclonal CD3 antibodies: a matter of timing. Am. J. Transplant. 12, 2909–2919 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Tuch, B. E., Hughes, T. C. & Evans, M. D. Encapsulated pancreatic progenitors derived from human embryonic stem cells as a therapy for insulin-dependent diabetes. Diabetes Metab. Res. Rev. 27, 928–932 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Zhi, Z. L., Khan, F. & Pickup, J. C. Multilayer nanoencapsulation: a nanomedicine technology for diabetes research and management. Diabetes Res. Clin. Pract. 100, 162–169 (2012).

    Article  PubMed  Google Scholar 

  21. Zhou, Q., Brown, J., Kanarek, A., Rajagopal, J. & Melton, D. A. In vivo reprogramming of adult pancreatic exocrine cells to β-cells. Nature 455, 627–632 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Thorel, F. et al. Conversion of adult pancreatic α-cells to β-cells after extreme β-cell loss. Nature 464, 1149–1154 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Jonsson, J., Carlsson, L., Edlund, T. & Edlund, H. Insulin-promoter-factor 1 is required for pancreas development in mice. Nature 371, 606–609 (1994).

    Article  CAS  PubMed  Google Scholar 

  24. Gradwohl, G., Dierich, A., LeMeur, M. & Guillemot, F. neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas. Proc. Natl Acad. Sci. USA 97, 1607–1611 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Gittes, G. K. Developmental biology of the pancreas: a comprehensive review. Dev. Biol. 326, 4–35 (2009).

    Article  CAS  PubMed  Google Scholar 

  26. Jensen, J. Gene regulatory factors in pancreatic development. Dev. Dyn. 229, 176–200 (2004).

    Article  CAS  PubMed  Google Scholar 

  27. Kim, S. K. & Hebrok, M. Intercellular signals regulating pancreas development and function. Genes Dev. 15, 111–127 (2001).

    Article  CAS  PubMed  Google Scholar 

  28. Oliver-Krasinski, J. M. & Stoffers, D. A. On the origin of the β cell. Genes Dev. 22, 1998–2021 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Bhushan, A. et al. Fgf10 is essential for maintaining the proliferative capacity of epithelial progenitor cells during early pancreatic organogenesis. Development 128, 5109–5117 (2001).

    CAS  PubMed  Google Scholar 

  30. Scharfmann, R. Control of early development of the pancreas in rodents and humans: implications of signals from the mesenchyme. Diabetologia 43, 1083–1092 (2000).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  32. Evans, M. J. & Kaufman, M. H. Establishment in culture of pluripotential cells from mouse embryos. Nature 292, 154–156 (1981).

    Article  CAS  PubMed  Google Scholar 

  33. Martin, G. R. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl Acad. Sci. USA 78, 7634–7638 (1981).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 1145–1147 (1998).

    Article  CAS  PubMed  Google Scholar 

  35. Lavon, N., Yanuka, O. & Benvenisty, N. The effect of overexpression of Pdx1 and Foxa2 on the differentiation of human embryonic stem cells into pancreatic cells. Stem Cells 24, 1923–1930 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Xu, H. et al. The combined expression of Pdx1 and MafA with either Ngn3 or NeuroD improve the differentiation efficiency of mouse embryonic stem cells into insulin-producing cells. Cell Transplant. 22, 147–158 (2013).

    Article  PubMed  Google Scholar 

  37. Hori, Y. et al. Growth inhibitors promote differentiation of insulin-producing tissue from embryonic stem cells. Proc. Natl Acad. Sci. USA 99, 16105–16110 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lumelsky, N. et al. Differentiation of embryonic stem cells to insulin-secreting structures similar to pancreatic islets. Science 292, 1389–1394 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Hansson, M. et al. Artifactual insulin release from differentiated embryonic stem cells. Diabetes 53, 2603–2609 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Rajagopal, J., Anderson, W. J., Kume, S., Martinez, O. I. & Melton, D. A. Insulin staining of ES cell progeny from insulin uptake. Science 299, 363 (2003).

    PubMed  Google Scholar 

  41. Mfopou, J. K., Chen, B., Sui, L., Sermon, K. & Bouwens, L. Recent advances and prospects in the differentiation of pancreatic cells from human embryonic stem cells. Diabetes 59, 2094–2101 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Cai, J. et al. Generation of homogeneous PDX1+ pancreatic progenitors from human ES cell-derived endoderm cells. J. Mol. Cell Biol. 2, 50–60 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  44. Mfopou, J. K., Chen, B., Mateizel, I., Sermon, K. & Bouwens, L. Noggin, retinoids, and fibroblast growth factor regulate hepatic or pancreatic fate of human embryonic stem cells. Gastroenterology 138, 2233–2245 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. 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  CAS  PubMed  PubMed Central  Google Scholar 

  47. 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  PubMed  Google Scholar 

  48. 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  PubMed  Google Scholar 

  49. 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  PubMed  PubMed Central  Google Scholar 

  50. Sui, L., Mfopou, J. K., Chen, B., Sermon, K. & Bouwens, L. Transplantation of human embryonic stem cell-derived pancreatic endoderm reveals a site-specific survival, growth and differentiation. Cell Transplant. 22, 821–830 (2013).

    Article  PubMed  Google Scholar 

  51. Cheng, X. et al. Self-renewing endodermal progenitor lines generated from human pluripotent stem cells. Cell Stem Cell 10, 371–384 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Sneddon, J. B., Borowiak, M. & Melton, D. A. Self-renewal of embryonic-stem-cell-derived progenitors by organ-matched mesenchyme. Nature 491, 765–768 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Sui, L., Geens, M., Sermon, K., Bouwens, L. & Mfopou, J. K. Role of BMP signaling in pancreatic progenitor differentiation from human embryonic stem cells. Stem Cell Rev. http://dx.doi.org/10.1007/s12015-013-9435-6.

  54. 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  PubMed  PubMed Central  Google Scholar 

  55. Richards, M., Fong, C. Y., Chan, W. K., Wong, P. C. & Bongso, A. Human feeders support prolonged undifferentiated growth of human inner cell masses and embryonic stem cells. Nat. Biotechnol. 20, 933–936 (2002).

    Article  CAS  PubMed  Google Scholar 

  56. Singh, A. M. et al. Signaling network crosstalk in human pluripotent cells: a Smad2/3-regulated switch that controls the balance between self-renewal and differentiation. Cell Stem Cell 10, 312–326 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Kunisada, Y., Tsubooka-Yamazoe, N., Shoji, M. & Hosoya, M. Small molecules induce efficient differentiation into insulin-producing cells from human induced pluripotent stem cells. Stem Cell Res. 8, 274–284 (2012).

    Article  CAS  PubMed  Google Scholar 

  58. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    Article  CAS  PubMed  Google Scholar 

  59. Zhang, D. et al. Highly efficient differentiation of human ES cells and iPS cells into mature pancreatic insulin-producing cells. Cell Res. 19, 429–438 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Chou, B. K. et al. Efficient human iPS cell derivation by a non-integrating plasmid from blood cells with unique epigenetic and gene expression signatures. Cell Res. 21, 518–529 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Okita, K. et al. An efficient nonviral method to generate integration-free human-induced pluripotent stem cells from cord blood and peripheral blood cells. Stem Cells 31, 458–466 (2013).

    Article  CAS  PubMed  Google Scholar 

  62. Puri, M. C. & Nagy, A. Concise review: embryonic stem cells versus induced pluripotent stem cells: the game is on. Stem Cells 30, 10–14 (2012).

    Article  CAS  PubMed  Google Scholar 

  63. Dor, Y., Brown, J., Martinez, O. I. & Melton, D. A. Adult pancreatic β-cells are formed by self-duplication rather than stem-cell differentiation. Nature 429, 41–46 (2004).

    Article  CAS  PubMed  Google Scholar 

  64. Nakamura, K. et al. Pancreatic β-cells are generated by neogenesis from non-β-cells after birth. Biomed. Res. 32, 167–174 (2011).

    Article  CAS  PubMed  Google Scholar 

  65. Teta, M., Rankin, M. M., Long, S. Y., Stein, G. M. & Kushner, J. A. Growth and regeneration of adult β cells does not involve specialized progenitors. Dev. Cell 12, 817–826 (2007).

    Article  CAS  PubMed  Google Scholar 

  66. Desai, B. M. et al. Preexisting pancreatic acinar cells contribute to acinar cell, but not islet β cell, regeneration. J. Clin. Invest. 117, 971–977 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kopinke, D. & Murtaugh, L. C. Exocrine-to-endocrine differentiation is detectable only prior to birth in the uninjured mouse pancreas. BMC Dev. Biol. 10, 38 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kopp, J. L. et al. Sox9+ ductal cells are multipotent progenitors throughout development but do not produce new endocrine cells in the normal or injured adult pancreas. Development 138, 653–665 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Kopp, J. L. et al. Identification of Sox9-dependent acinar-to-ductal reprogramming as the principal mechanism for initiation of pancreatic ductal adenocarcinoma. Cancer Cell 22, 737–750 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Pan, F. C. et al. Spatiotemporal patterns of multipotentiality in Ptf1a-expressing cells during pancreas organogenesis and injury-induced facultative restoration. Development 140, 751–764 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Solar, M. et al. Pancreatic exocrine duct cells give rise to insulin-producing β cells during embryogenesis but not after birth. Dev. Cell 17, 849–860 (2009).

    Article  CAS  PubMed  Google Scholar 

  72. Inada, A. et al. Carbonic anhydrase II-positive pancreatic cells are progenitors for both endocrine and exocrine pancreas after birth. Proc. Natl Acad. Sci. USA 105, 19915–19919 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Wang, R. N., Kloppel, G. & Bouwens, L. Duct- to islet-cell differentiation and islet growth in the pancreas of duct-ligated adult rats. Diabetologia 38, 1405–1411 (1995).

    Article  CAS  PubMed  Google Scholar 

  74. Erickson, R. P., Grimes, J., Venta, P. J. & Tashian, R. E. Expression of carbonic anhydrase II (CA II) promoter-reporter fusion genes in multiple tissues of transgenic mice does not replicate normal patterns of expression indicating complexity of CA II regulation in vivo. Biochem. Genet. 33, 421–437 (1995).

    Article  CAS  PubMed  Google Scholar 

  75. Furuyama, K. et al. Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat. Genet. 43, 34–41 (2011).

    Article  CAS  PubMed  Google Scholar 

  76. Kopp, J. L. et al. Progenitor cell domains in the developing and adult pancreas. Cell Cycle 10, 1921–1927 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Xu, X. et al. β cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell 132, 197–207 (2008).

    Article  CAS  PubMed  Google Scholar 

  78. Rovira, M. et al. Isolation and characterization of centroacinar/terminal ductal progenitor cells in adult mouse pancreas. Proc. Natl Acad. Sci. USA 107, 75–80 (2010).

    Article  CAS  PubMed  Google Scholar 

  79. Seaberg, R. M. et al. Clonal identification of multipotent precursors from adult mouse pancreas that generate neural and pancreatic lineages. Nat. Biotechnol. 22, 1115–1124 (2004).

    Article  CAS  PubMed  Google Scholar 

  80. Smukler, S. R. et al. The adult mouse and human pancreas contain rare multipotent stem cells that express insulin. Cell Stem Cell 8, 281–293 (2011).

    Article  CAS  PubMed  Google Scholar 

  81. Kopinke, D. et al. Lineage tracing reveals the dynamic contribution of Hes1+ cells to the developing and adult pancreas. Development 138, 431–441 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Russ, H. A., Bar, Y., Ravassard, P. & Efrat, S. In vitro proliferation of cells derived from adult human β-cells revealed by cell-lineage tracing. Diabetes 57, 1575–1583 (2008).

    Article  CAS  PubMed  Google Scholar 

  83. Russ, H. A. et al. Insulin-producing cells generated from dedifferentiated human pancreatic β cells expanded in vitro. PLoS ONE 6, e25566 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Bar, Y. et al. Redifferentiation of expanded human pancreatic β-cell-derived cells by inhibition of the NOTCH pathway. J. Biol. Chem. 287, 17269–17280 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Atouf, F. et al. No evidence for mouse pancreatic β-cell epithelial-mesenchymal transition in vitro. Diabetes 56, 699–702 (2007).

    Article  CAS  PubMed  Google Scholar 

  86. Chase, L. G., Ulloa-Montoya, F., Kidder, B. L. & Verfaillie, C. M. Islet-derived fibroblast-like cells are not derived via epithelial-mesenchymal transition from Pdx-1 or insulin-positive cells. Diabetes 56, 3–7 (2007).

    Article  CAS  PubMed  Google Scholar 

  87. Morton, R. A., Geras-Raaka, E., Wilson, L. M., Raaka, B. M. & Gershengorn, M. C. Endocrine precursor cells from mouse islets are not generated by epithelial-to-mesenchymal transition of mature β cells. Mol. Cell Endocrinol. 270, 87–93 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Weinberg, N., Ouziel-Yahalom, L., Knoller, S., Efrat, S. & Dor, Y. Lineage tracing evidence for in vitro dedifferentiation but rare proliferation of mouse pancreatic β-cells. Diabetes 56, 1299–1304 (2007).

    Article  CAS  PubMed  Google Scholar 

  89. Talchai, C., Xuan, S., Lin, H. V., Sussel, L. & Accili, D. Pancreatic β-cell dedifferentiation as a mechanism of diabetic β-cell failure. Cell 150, 1223–1234 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Houbracken, I. & Bouwens, L. The quest for tissue stem cells in the pancreas and other organs, and their application in β-cell replacement. Rev. Diabet. Stud. 7, 112–123 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  91. Nombela-Arrieta, C., Ritz, J. & Silberstein, L. E. The elusive nature and function of mesenchymal stem cells. Nat. Rev. Mol. Cell Biol. 12, 126–131 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Lee, V. M. & Stoffel, M. Bone marrow: an extra-pancreatic hideout for the elusive pancreatic stem cell? J. Clin. Invest. 111, 799–801 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Ianus, A., Holz, G. G., Theise, N. D. & Hussain, M. A. In vivo derivation of glucose-competent pancreatic endocrine cells from bone marrow without evidence of cell fusion. J. Clin. Invest. 111, 843–850 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Taneera, J. et al. Failure of transplanted bone marrow cells to adopt a pancreatic β-cell fate. Diabetes 55, 290–296 (2006).

    Article  CAS  PubMed  Google Scholar 

  95. Jiang, Y. et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 418, 41–49 (2002).

    Article  CAS  PubMed  Google Scholar 

  96. Kodama, S., Kuhtreiber, W., Fujimura, S., Dale, E. A. & Faustman, D. L. Islet regeneration during the reversal of autoimmune diabetes in NOD mice. Science 302, 1223–1227 (2003).

    Article  CAS  PubMed  Google Scholar 

  97. Couzin, J. Immunology. Diabetes studies conflict on power of spleen cells. Science 311, 1694 (2006).

    Article  CAS  PubMed  Google Scholar 

  98. Mathews, V. et al. Recruitment of bone marrow-derived endothelial cells to sites of pancreatic β-cell injury. Diabetes 53, 91–98 (2004).

    Article  CAS  PubMed  Google Scholar 

  99. Bell, G. I. et al. Transplanted human bone marrow progenitor subtypes stimulate endogenous islet regeneration and revascularization. Stem Cells Dev. 21, 97–109 (2012).

    Article  CAS  PubMed  Google Scholar 

  100. Hess, D. et al. Bone marrow-derived stem cells initiate pancreatic regeneration. Nat. Biotechnol. 21, 763–770 (2003).

    Article  CAS  PubMed  Google Scholar 

  101. Fiorina, P., Voltarelli, J. & Zavazava, N. Immunological applications of stem cells in type 1 diabetes. Endocr. Rev. 32, 725–754 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Nicholas, C. R. & Kriegstein, A. R. Regenerative medicine: cell reprogramming gets direct. Nature 463, 1031–1032 (2010).

    Article  CAS  PubMed  Google Scholar 

  103. 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  PubMed  PubMed Central  Google Scholar 

  104. Sapir, T. et al. Cell-replacement therapy for diabetes: generating functional insulin-producing tissue from adult human liver cells. Proc. Natl Acad. Sci. USA 102, 7964–7969 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Lima, M. J. et al. Suppression of epithelial to mesenchymal transitioning (EMT) enhances ex vivo reprogramming of human exocrine pancreatic tissue towards functional insulin producing β-like cells. Diabetes http://dx.doi.org/10.2337/db12-1256.

  106. Baeyens, L. et al. In vitro generation of insulin-producing β cells from adult exocrine pancreatic cells. Diabetologia 48, 49–57 (2005).

    Article  CAS  PubMed  Google Scholar 

  107. Minami, K. et al. Lineage tracing and characterization of insulin-secreting cells generated from adult pancreatic acinar cells. Proc. Natl Acad. Sci. USA 102, 15116–15121 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Baeyens, L. et al. Notch signaling as gatekeeper of rat acinar-to-β-cell conversion in vitro. Gastroenterology 136, 1750–1760.e13 (2009).

    Article  CAS  PubMed  Google Scholar 

  109. Pinho, A. V. et al. Adult pancreatic acinar cells dedifferentiate to an embryonic progenitor phenotype with concomitant activation of a senescence programme that is present in chronic pancreatitis. Gut 60, 958–966 (2011).

    Article  CAS  PubMed  Google Scholar 

  110. Lardon, J. et al. Plasticity in the adult rat pancreas: transdifferentiation of exocrine to hepatocyte-like cells in primary culture. Hepatology 39, 1499–1507 (2004).

    Article  CAS  PubMed  Google Scholar 

  111. Hansen, J. B. et al. Divalent metal transporter 1 regulates iron-mediated ROS and pancreatic β-cell fate in response to cytokines. Cell Metab. 16, 449–461 (2012).

    Article  CAS  PubMed  Google Scholar 

  112. Houbracken, I. et al. Lineage tracing evidence for transdifferentiation of acinar to duct cells and plasticity of human pancreas. Gastroenterology 141, 731–741 (2011).

    Article  PubMed  Google Scholar 

  113. Collombat, P. et al. The ectopic expression of Pax4 in the mouse pancreas converts progenitor cells into α and subsequently β cells. Cell 138, 449–462 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Bramswig, N. C. et al. Epigenomic plasticity enables human pancreatic α to β cell reprogramming. J. Clin. Invest. 123, 1275–1284 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Lanza, R. P., Hayes, J. L. & Chick, W. L. Encapsulated cell technology. Nat. Biotechnol. 14, 1107–1111 (1996).

    Article  CAS  PubMed  Google Scholar 

  116. Dean, S. K., Yulyana, Y., Williams, G., Sidhu, K. S. & Tuch, B. E. Differentiation of encapsulated embryonic stem cells after transplantation. Transplantation 82, 1175–1184 (2006).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors' research was supported by the Research Foundation Flanders (FWO) and the European Union (FP7).

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of this article.

Corresponding author

Correspondence to Luc Bouwens.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bouwens, L., Houbracken, I. & Mfopou, J. The use of stem cells for pancreatic regeneration in diabetes mellitus. Nat Rev Endocrinol 9, 598–606 (2013). https://doi.org/10.1038/nrendo.2013.145

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrendo.2013.145

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