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:

Regenerative medicine and cell-based approaches to restore pancreatic function

Key Points

  • Both exocrine and endocrine pancreatic pathologies bear tremendous costs to health-care systems and patients alike; identifying cell-based therapies for restoring exocrine pancreatic insufficiency or replacing inadequate β cells in patients with diabetes are important targets

  • Two strategies for restoring pancreatic function are through differentiation of stem cells and reprogramming of cells through gene therapy

  • Beyond functional assessments, considerations for delivery of stem-cell-derived β cells include implant site and decreasing immunosuppression requirements

  • In situ reprogramming strategies must consider different risks than cell delivery approaches, including lack of specificity and unpredictable off-target effects

  • Neither approach is ready to supersede enzyme replacement therapy or exogenous insulin delivery, but with continued progress cell replacement strategies could become the standard of care

Abstract

The pancreas is a complex organ with exocrine and endocrine components. Many pathologies impair exocrine function, including chronic pancreatitis, cystic fibrosis and pancreatic ductal adenocarcinoma. Conversely, when the endocrine pancreas fails to secrete sufficient insulin, patients develop diabetes mellitus. Pathology in either the endocrine or exocrine pancreas results in devastating economic and personal consequences. The current standard therapy for treating patients with type 1 diabetes mellitus is daily exogenous insulin injections, but cell sources of insulin provide superior glycaemic regulation and research is now focused on the goal of regenerating or replacing β cells. Stem-cell-based models might be useful to study exocrine pancreatic disorders, and mesenchymal stem cells or secreted factors might delay disease progression. Although the standards that bioengineered cells must meet before being considered as a viable therapy are not yet established, any potential therapy must be acceptably safe and functionally superior to current therapies. Here, we describe progress and challenges in cell-based methods to restore pancreatic function, with a focus on optimizing the site for cell delivery and decreasing requirements for immunosuppression through encapsulation. We also discuss the tools and strategies being used to generate exocrine pancreas and insulin-producing β-cell surrogates in situ and highlight obstacles to clinical application.

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: The healthy human pancreas contains both an endocrine and an exocrine compartment.
Figure 2: Examples of endocrine and exocrine pancreatic pathologies.

Similar content being viewed by others

Zixuan Zhao, Xinyi Chen, … Hanry Yu

References

  1. Taylor, K. W. Pathogenesis of diabetes mellitus. J. Clin. Pathol. Suppl. (Assoc. Clin. Pathol.) 2, 76–81 (1969).

    Article  Google Scholar 

  2. Fieker, A., Philpott, J. & Armand, M. Enzyme replacement therapy for pancreatic insufficiency: present and future. Clin. Exp. Gastroenterol. 4, 55–73 (2011).

    PubMed  PubMed Central  Google Scholar 

  3. Piciucchi, M. et al. Exocrine pancreatic insufficiency in diabetic patients: prevalence, mechanisms, and treatment. Int. J. Endocrinol. 2015, 595649 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. 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  PubMed  Google Scholar 

  5. Kieffer, T. J. Closing in on mass production of mature human beta cells. Cell Stem Cell 18, 699–702 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Johnson, J. D. The quest to make fully functional human pancreatic beta cells from embryonic stem cells: climbing a mountain in the clouds. Diabetologia 59, 2047–2057 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Zulewski, H. et al. Multipotential nestin-positive stem cells isolated from adult pancreatic islets differentiate ex vivo into pancreatic endocrine, exocrine, and hepatic phenotypes. Diabetes 50, 521–533 (2001).

    Article  CAS  PubMed  Google Scholar 

  8. Delaspre, F. et al. Directed pancreatic acinar differentiation of mouse embryonic stem cells via embryonic signalling molecules and exocrine transcription factors. PLoS ONE 8, e54243 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Stern, R. C., Mayes, J. T., Weber, F. L., Blades, E. W. & Schulak, J. A. Restoration of exocrine pancreatic function following pancreas–liver–kidney transplantation in a cystic fibrosis patient. Clin. Transplant. 8, 1–4 (1994).

    CAS  PubMed  Google Scholar 

  10. Gruessner, R. W. G., Manivel, C., Dunn, D. L. & Sutherland, D. E. R. Pancreaticoduodenal transplantation with enteric drainage following native total pancreatectomy for chronic pancreatitis: a case report. Pancreas 6, 479–488 (1991).

    Article  CAS  PubMed  Google Scholar 

  11. Usatin, D. J., Perito, E. R., Posselt, A. M. & Rosenthal, P. Under utilization of pancreas transplants in cystic fibrosis recipients in the United Network Organ Sharing (UNOS) Data 1987–2014. Am. J. Transplant. 16, 1620–1625 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Chen, Z., Lu, F., Fang, H. & Huang, H. Effect of mesenchymal stem cells on renal injury in rats with severe acute pancreatitis. Exp. Biol. Med. (Maywood) 238, 687–695 (2013).

    Article  CAS  Google Scholar 

  13. Tu, X. H. et al. Role of bone marrow-derived mesenchymal stem cells in a rat model of severe acute pancreatitis. World J. Gastroenterol. 18, 2270–2279 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Cui, H. F. & Bai, Z. L. Protective effects of transplanted and mobilized bone marrow stem cells on mice with severe acute pancreatitis. World J. Gastroenterol. 9, 2274–2277 (2003).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Jung, K. H. et al. Human bone marrow-derived clonal mesenchymal stem cells inhibit inflammation and reduce acute pancreatitis in rats. Gastroenterology 140, 998–1008 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Yang, B. et al. Effect of umbilical cord mesenchymal stem cells on treatment of severe acute pancreatitis in rats. Cytotherapy 15, 154–162 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Yin, G. et al. Role of bone marrow mesenchymal stem cells in L-arg-induced acute pancreatitis: effects and possible mechanisms. Int. J. Clin. Exp. Pathol. 8, 4457–4468 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Murtaugh, L. C. & Keefe, M. Regeneration and repair of the exocrine pancreas. Annu. Rev. Physiol. 77, 229–249 (2015).

    Article  CAS  PubMed  Google Scholar 

  19. Maxmen, A. Exome sequencing deciphers rare diseases. Cell 144, 635–637 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Tulpule, A. et al. Pluripotent stem cell models of Shwachman–Diamond syndrome reveal a common mechanism for pancreatic and hematopoietic dysfunction. Cell Stem Cell 12, 727–736 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hohwieler, M. et al. Human pluripotent stem cell-derived acinar/ductal organoids generate human pancreas upon orthotopic transplantation and allow disease modelling. Gut 66, 473–486 (2017).

    Article  CAS  PubMed  Google Scholar 

  22. Dalla Pozza, E. et al. Pancreatic ductal adenocarcinoma cell lines display a plastic ability to bidirectionally convert into cancer stem cells. Int. J. Oncol. 46, 1099–1108 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Michalopoulos, G. K. Liver regeneration. J. Cell. Physiol. 213, 286–300 (2007).]

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Beer, R. L., Parsons, M. J. & Rovira, M. Centroacinar cells: at the center of pancreas regeneration. Dev. Biol. 413, 8–15 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kopp, J. L., Grompe, M. & Sander, M. Stem cells versus plasticity in liver and pancreas regeneration. Nat. Cell Biol. 18, 238–245 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Menge, B. A. et al. Partial pancreatectomy in adult humans does not provoke β-cell regeneration. Diabetes 57, 142–149 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Tsiotos, G. G., Barry, M. K., Johnson, C. D. & Sarr, M. G. Pancreas regeneration after resection: does it occur in humans? Pancreas 19, 310–313 (1999).

    Article  CAS  PubMed  Google Scholar 

  28. Berrocal, T., Luque, A. A., Pinilla, I. & Lassaletta, L. Pancreatic regeneration after near-total pancreatectomy in children with nesidioblastosis. Pediatr. Radiol. 35, 1066–1070 (2005).

    Article  PubMed  Google Scholar 

  29. Risbud, M. V. & Bhonde, R. R. Models of pancreatic regeneration in diabetes. Diabetes Res. Clin. Pract. 58, 155–165 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. 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). This study gives insight into the progression of pancreatic ductal adenocarcinoma and might provide a target for future treatment.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Parsons, M. J. et al. Notch-responsive cells initiate the secondary transition in larval zebrafish pancreas. Mech. Dev. 126, 898–912 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Delaspre, F. et al. Centroacinar cells are progenitors that contribute to endocrine pancreas regeneration. Diabetes 64, 3499–3509 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. 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 

  34. 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 

  35. Stanger, B. Z. et al. Pten constrains centroacinar cell expansion and malignant transformation in the pancreas. Cancer Cell 8, 185–195 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. 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 

  37. Van de Casteele, M. et al. Partial duct ligation: beta-cell proliferation and beyond. Diabetes 63, 2567–2577 (2014).

    Article  CAS  PubMed  Google Scholar 

  38. Van de Casteele, M. et al. Neurogenin 3+ cells contribute to beta-cell neogenesis and proliferation in injured adult mouse pancreas. Cell Death Dis. 4, e523 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Maeda, H., Danel, C. & Crystal, R. G. Adenovirus-mediated transfer of human lipase complementary DNA to the gallbladder. Gastroenterology 106, 1638–1644 (1994).

    Article  CAS  PubMed  Google Scholar 

  40. Kuhel, D. G., Zheng, S., Tso, P. & Hui, D. Y. Adenovirus-mediated human pancreatic lipase gene transfer to rat bile: gene therapy of fat malabsorption. Am. J. Physiol. Gastrointest. Liver Physiol. 279, G1031–G1036 (2000).

    Article  CAS  PubMed  Google Scholar 

  41. Houbracken, I., Baeyens, L., Ravassard, P., Heimberg, H. & Bouwens, L. Gene delivery to pancreatic exocrine cells in vivo and in vitro. BMC Biotechnol. 12, 74 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Shapiro, A. M. J., Pokrywczynska, M. & Ricordi, C. Clinical pancreatic islet transplantation. Nat. Rev. Endocrinol. 13, 268–277 (2016).

    Article  CAS  PubMed  Google Scholar 

  43. Bruni, A., Gala-Lopez, B., Pepper, A. R., Abualhassan, N. S. & Shapiro, A. M. J. Islet cell transplantation for the treatment of type 1 diabetes: recent advances and future challenges. Diabetes Metab. Syndr. Obes. 7, 211–223 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Ricordi, C. et al. National Institutes of Health-sponsored Clinical Islet Transplantation Consortium phase 3 trial: manufacture of a complex cellular product at eight processing facilities. Diabetes 65, 3418–3428 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hering, B. J. et al. Phase 3 trial of transplantation of human islets in type 1 diabetes complicated by severe hypoglycemia. Diabetes Care 39, 1230–1240 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hathout, E., Lakey, J. & Shapiro, J. Islet transplant: an option for childhood diabetes? Arch. Dis. Child. 88, 591–594 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Bennet, W., Groth, C. G., Larsson, R., Nilsson, B. & Korsgren, O. Isolated human islets trigger an instant blood mediated inflammatory reaction: implications for intraportal islet transplantation as a treatment for patients with type 1 diabetes. Ups. J. Med. Sci. 105, 125–133 (2000).

    Article  CAS  PubMed  Google Scholar 

  48. Moberg, L. et al. Production of tissue factor by pancreatic islet cells as a trigger of detrimental thrombotic reactions in clinical islet transplantation. Lancet 360, 2039–2045 (2002).

    Article  CAS  PubMed  Google Scholar 

  49. Drachenberg, C. B. et al. Islet cell damage associated with tacrolimus and cyclosporine: morphological features in pancreas allograft biopsies and clinical correlation. Transplantation 68, 396–402 (1999).

    Article  CAS  PubMed  Google Scholar 

  50. Ellis, C. E. & Korbutt, G. S. Justifying clinical trials for porcine islet xenotransplantation. Xenotransplantation 22, 336–344 (2015).

    Article  PubMed  Google Scholar 

  51. Cardona, K. et al. Long-term survival of neonatal porcine islets in nonhuman primates by targeting costimulation pathways. Nat. Med. 12, 304–306 (2006). This preclinical work provides evidence that neonatal porcine islets are a potential source of islets to treat human patients with diabetes.

    Article  CAS  PubMed  Google Scholar 

  52. Hering, B. J. et al. Prolonged diabetes reversal after intraportal xenotransplantation of wild-type porcine islets in immunosuppressed nonhuman primates. Nat. Med. 12, 301–303 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. van der Windt, D. J. et al. Clinical islet xenotransplantation. How close are we? Diabetes 61, 3046–3055 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Samy, K. P., Martin, B. M., Turgeon, N. A. & Kirk, A. D. Islet cell xenotransplantation: a serious look toward the clinic. Xenotransplantation 21, 221–229 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  55. van der Laan, L. J. et al. Infection by porcine endogenous retrovirus after islet xenotransplantation in SCID mice. Nature 407, 90–94 (2000).

    Article  CAS  PubMed  Google Scholar 

  56. Reardon, S. New life for pig-to-human transplants. Nature 527, 152–154 (2015).

    Article  CAS  PubMed  Google Scholar 

  57. Yang, L. et al. Genome-wide inactivation of porcine endogenous retroviruses (PERVs). Science 350, 1101–1104 (2015).

    Article  CAS  PubMed  Google Scholar 

  58. Reyes, L. M. et al. Creating class I MHC-null pigs using guide RNA and the Cas9 endonuclease. J. Immunol. 193, 5751–5757 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Dai, Y. et al. Targeted disruption of the α1,3-galactosyltransferase gene in cloned pigs. Nat. Biotechnol. 20, 251–255 (2002).

    Article  CAS  PubMed  Google Scholar 

  60. Lai, L. et al. Production of alpha-1,3-galactosyltransferase knockout pigs by nuclear transfer cloning. Science 295, 1089–1092 (2002).

    Article  CAS  PubMed  Google Scholar 

  61. Yamada, K. et al. Marked prolongation of porcine renal xenograft survival in baboons through the use of α1,3-galactosyltransferase gene-knockout donors and the cotransplantation of vascularized thymic tissue. Nat. Med. 11, 32–34 (2005).

    Article  CAS  PubMed  Google Scholar 

  62. Nagashima, H. & Matsunari, H. Growing human organs in pigs — a dream or reality? Theriogenology 86, 422–426 (2016).

    Article  PubMed  Google Scholar 

  63. Kobayashi, T. et al. Generation of rat pancreas in mouse by interspecific blastocyst injection of pluripotent stem cells. Cell 142, 787–799 (2010).

    Article  CAS  PubMed  Google Scholar 

  64. Yamaguchi, T. et al. Interspecies organogenesis generates autologous functional islets. Nature 542, 191–196 (2017). This study provides proof-of concept that pluripotent stem cell derived islets can be generated in a xenogeneic host.

    Article  CAS  PubMed  Google Scholar 

  65. Matsunari, H. et al. Blastocyst complementation generates exogenic pancreas in vivo in apancreatic cloned pigs. Proc. Natl Acad. Sci. USA 110, 4557–4562 (2013).

    Article  PubMed  Google Scholar 

  66. Faradji, R. N. et al. Glucose-induced toxicity in insulin-producing pituitary cells that coexpress GLUT2 and glucokinase. Implications for metabolic engineering. J. Biol. Chem. 276, 36695–36702 (2001).

    Article  CAS  PubMed  Google Scholar 

  67. Xie, M. et al. beta-cell-mimetic designer cells provide closed-loop glycemic control. Science 354, 1296–1301 (2016).

    Article  CAS  PubMed  Google Scholar 

  68. Hohmeier, H. E. et al. Isolation of INS-1-derived cell lines with robust ATP-sensitive K+ channel-dependent and -independent glucose-stimulated insulin secretion. Diabetes 49, 424–430 (2000).

    Article  CAS  PubMed  Google Scholar 

  69. Clark, S. A. et al. Novel insulinoma cell lines produced by iterative engineering of GLUT2, glucokinase, and human insulin expression. Diabetes 46, 958–967 (1997).

    Article  CAS  PubMed  Google Scholar 

  70. Ravassard, P. et al. A genetically engineered human pancreatic beta cell line exhibiting glucose-inducible insulin secretion. J. Clin. Invest. 121, 3589–3597 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Scharfmann, R. et al. Development of a conditionally immortalized human pancreatic beta cell line. J. Clin. Invest. 124, 2087–2098 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Murry, C. E. & Keller, G. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell 132, 661–680 (2008).

    Article  CAS  PubMed  Google Scholar 

  73. MacDonald, P. E., Joseph, J. W. & Rorsman, P. Glucose-sensing mechanisms in pancreatic β-cells. Phil. Trans. R. Soc. Lond. B Biol. Sci. 360, 2211–2225 (2005).

    Article  CAS  Google Scholar 

  74. Castaing, M. et al. Blood glucose normalization upon transplantation of human embryonic pancreas into beta-cell-deficient SCID mice. Diabetologia 44, 2066–2076 (2001).

    Article  CAS  PubMed  Google Scholar 

  75. Tuch, B. E., Sheil, A. G., Ng, A. B., Trent, R. J. & Turtle, J. R. Recovery of human fetal pancreas after one year of implantation in the diabetic patient. Transplantation 46, 865–870 (1988).

    Article  CAS  PubMed  Google Scholar 

  76. Lafferty, K. J. & Hao, L. Fetal pancreas transplantation for treatment of IDDM patients. Diabetes Care 16, 383–386 (1993).

    Article  CAS  PubMed  Google Scholar 

  77. Farkas, G. & Karacsonyi, S. Clinical transplantation of fetal human pancreatic islets. Biomed. Biochim. Acta 44, 155–159 (1985).

    CAS  PubMed  Google Scholar 

  78. D'Amour, K. A. et al. Production of pancreatic hormone-expressing endocrine cells from human embryonic stem cells. Nat. Biotechnol. 24, 1392–1401 (2006). This work was among the first to demonstate that insulin-producing cells can be derived from human pluripotent stem cells.

    Article  CAS  PubMed  Google Scholar 

  79. 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). This work convincingly demonstrated that implant of stem-cell-derived pancreatic progenitor cells could reverse diabetes in rodents, providing impetus for ViaCyte to initiate clinical trials.

    Article  CAS  PubMed  Google Scholar 

  80. 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 

  81. 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  PubMed  Google Scholar 

  82. 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  PubMed  Google Scholar 

  83. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02239354 (2017).

  84. Quiskamp, N., Bruin, J. E. & Kieffer, T. J. Differentiation of human pluripotent stem cells into β-cells: potential and challenges. Best Pract. Res. Clin. Endocrinol. Metab. 29, 833–847 (2015).

    Article  CAS  PubMed  Google Scholar 

  85. Rezania, A. et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat. Biotechnol. 32, 1121–1133 (2014). This manuscript describes a protocol to efficiently differentiate human embryonic stem cells and induced pluripotent stem cells into glucose-reseponsive insulin-producing cells resembling immature β cells that reversed diabetes in mice quicker than pancreatic progenitor cells.

    Article  CAS  PubMed  Google Scholar 

  86. Pagliuca, F. W. et al. Generation of functional human pancreatic beta cells in vitro. Cell 159, 428–439 (2014). In this paper, the authors describe the differentiation of human pluripotent stem cells into cells with key characteristics of β cells that were able to prevent diabetes onset in a mouse model.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Russ, H. A. et al. Controlled induction of human pancreatic progenitors produces functional beta-like cells in vitro. EMBO J. 34, 1759–1772 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006). This was the first manuscript to describe methods for the generation of induced pluripotent stem cells, work that eventually earned Yamanaka a share of the 2012 Nobel Prize in Physiology or Medicine.

    Article  CAS  PubMed  Google Scholar 

  89. Zhu, S. et al. Human pancreatic beta-like cells converted from fibroblasts. Nat. Commun. 7, 10080 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Cyranoski, D. Japanese man is first to receive 'reprogrammed' stem cells from another person. Nature http://dx.doi.org/10.1038/nature.2017.21730 (2017).

  91. Medawar, P. Immunity to homologous grafted skin. III. The fate of skin homografts transplanted to the brain to subcutaneous tissue, and to the anterior chamber of the eye. Br. J. Exp. Pathol. 29, 58–69 (1948).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. van der Torren, C. R. et al. Immunogenicity of human embryonic stem cell-derived beta cells. Diabetologia 60, 126–133 (2017).

    Article  CAS  PubMed  Google Scholar 

  93. Riolobos, L. et al. HLA engineering of human pluripotent stem cells. Mol. Ther. 21, 1232–1241 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Figueiredo, C. & Blasczyk, R. A future with less HLA: potential clinical applications of HLA-universal cells. Tissue Antigens 85, 443–449 (2015).

    Article  CAS  PubMed  Google Scholar 

  95. Kent, S. C. et al. Expanded T cells from pancreatic lymph nodes of type 1 diabetic subjects recognize an insulin epitope. Nature 435, 224–228 (2005).

    Article  CAS  PubMed  Google Scholar 

  96. Pepper, A. R., Gala-Lopez, B., Ziff, O. & Shapiro, A. M. Revascularization of transplanted pancreatic islets and role of the transplantation site. Clin. Dev. Immunol. 2013, 352315 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Nanji, S. A. & Shapiro, A. M. J. Advances in pancreatic islet transplantation in humans. Diabetes Obes. Metab. 8, 15–25 (2006).

    Article  PubMed  Google Scholar 

  98. Villiger, P. et al. Prevention of bleeding after islet transplantation: lessons learned from a multivariate analysis of 132 cases at a single institution. Am. J. Transplant. 5, 2992–2998 (2005).

    Article  CAS  PubMed  Google Scholar 

  99. Barshes, N. R. et al. Transaminitis after pancreatic islet transplantation. J. Am. Coll. Surg. 200, 353–361 (2005).

    Article  PubMed  Google Scholar 

  100. Markmann, J. F. et al. Magnetic resonance-defined periportal steatosis following intraportal islet transplantation — a functional footprint of islet graft survival? Diabetes 52, 1591–1594 (2003).

    Article  CAS  PubMed  Google Scholar 

  101. Bhargava, R. et al. Prevalence of hepatic steatosis after islet transplantation and its relation to graft function. Diabetes 53, 1311–1317 (2004).

    Article  CAS  PubMed  Google Scholar 

  102. Veriter, S., Gianello, P. & Dufrane, D. Bioengineered sites for islet cell transplantation. Curr. Diab. Rep. 13, 745–755 (2013).

    Article  CAS  PubMed  Google Scholar 

  103. Ferguson, J., Scothorne, R. J. & Johnston, I. D. Proceedings: the survival of transplanted isolated pancreatic islets in the omentum and testis. Br. J. Surg. 60, 907 (1973).

    CAS  PubMed  Google Scholar 

  104. Kriz, J. et al. A novel technique for the transplantation of pancreatic islets within a vascularized device into the greater omentum to achieve insulin independence. Am. J. Surg. 203, 793–797 (2012).

    Article  PubMed  Google Scholar 

  105. Kin, T., Korbutt, G. S. & Rajotte, R. V. Survival and metabolic function of syngeneic rat islet grafts transplanted in the omental pouch. Am. J. Transplant. 3, 281–285 (2003).

    Article  PubMed  Google Scholar 

  106. Berman, D. M. et al. Bioengineering the endocrine pancreas: intraomental islet transplantation within a biologic resorbable scaffold. Diabetes 65, 1350–1361 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02213003 (2017).

  108. Schmidt, S. Pancreatic islets find a new transplant home in the omentum. Nat. Biotechnol. 35, 8 (2017).

    Article  CAS  PubMed  Google Scholar 

  109. Caiazzo, R. et al. Evaluation of alternative sites for islet transplantation in the minipig: interest and limits of the gastric submucosa. Transplant. Proc. 39, 2620–2623 (2007).

    Article  CAS  PubMed  Google Scholar 

  110. Echeverri, G. J. et al. Endoscopic Gastric Submucosal Transplantation of Islets (ENDO-STI): technique and initial results in diabetic pigs. Am. J. Transplant. 9, 2485–2496 (2009).

    Article  CAS  PubMed  Google Scholar 

  111. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT01571817 (2013).

  112. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02402439 (2017).

  113. Tsuchiya, H. et al. Extracellular matrix and growth factors improve the efficacy of intramuscular islet transplantation. PLoS ONE 10, e0140910 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Christoffersson, G. et al. Clinical and experimental pancreatic islet transplantation to striated muscle: establishment of a vascular system similar to that in native islets. Diabetes 59, 2569–2578 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Christoffersson, G., Carlsson, P.-O. & Phillipson, M. Intramuscular islet transplantation promotes restored islet vascularity. Islets 3, 69–71 (2011).

    Article  PubMed  Google Scholar 

  116. Sterkers, A. et al. Islet survival and function following intramuscular autotransplantation in the minipig. Am. J. Transplant. 13, 891–898 (2013).

    Article  CAS  PubMed  Google Scholar 

  117. Garcea, G. et al. Total pancreatectomy with and without islet cell transplantation for chronic pancreatitis: a series of 85 consecutive patients. Pancreas 38, 1–7 (2009).

    Article  PubMed  Google Scholar 

  118. Webb, M. A. et al. Islet auto transplantation following total pancreatectomy: a long-term assessment of graft function. Pancreas 37, 282–287 (2008).

    Article  PubMed  Google Scholar 

  119. Rafael, E. et al. Intramuscular autotransplantation of pancreatic islets in a 7-year-old child: a 2-year follow-up. Am. J. Transplant. 8, 458–462 (2008).

    Article  CAS  PubMed  Google Scholar 

  120. Cantarelli, E. et al. Bone marrow as an alternative site for islet transplantation. Blood 114, 4566–4574 (2009).

    Article  CAS  PubMed  Google Scholar 

  121. Maffi, P. et al. Autologous pancreatic islet transplantation in human bone marrow. Diabetes 62, 3523–3531 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Stendahl, J. C., Kaufman, D. B. & Stupp, S. I. Extracellular matrix in pancreatic islets: relevance to scaffold design and transplantation. Cell Transplant. 18, 1–12 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Yuan, Y. et al. Self-assembling peptide nanofiber as potential substrates in islet transplantation. Transplant. Proc. 40, 2571–2574 (2008).

    Article  CAS  PubMed  Google Scholar 

  124. Cheng, J. Y. C., Raghunath, M., Whitelock, J. & Poole-Warren, L. Matrix components and scaffolds for sustained islet function. Tissue Eng. Part B Rev. 17, 235–247 (2011).

    Article  CAS  PubMed  Google Scholar 

  125. Golocheikine, A. et al. Cooperative signaling for angiogenesis and neovascularization by VEGF and HGF following islet transplantation. Transplantation 90, 725–731 (2010).

    Article  CAS  PubMed  Google Scholar 

  126. Jiwon, S. & Jeffrey, R. M. Economic 3D-printing approach for transplantation of human stem cell-derived β -like cells. Biofabrication 9, 015002 (2017).

    Google Scholar 

  127. Smink, A. M. et al. The efficacy of a prevascularized, retrievable poly(D,L,-lactide-co-ε-caprolactone) subcutaneous scaffold as transplantation site for pancreatic islets. Transplantation 101, e112–e119 (2017).

    Article  CAS  PubMed  Google Scholar 

  128. Pepper, A. R. et al. A prevascularized subcutaneous device-less site for islet and cellular transplantation. Nat. Biotechnol. 33, 518–523 (2015). This novel work showed that islets can survive following subcutaneous transplantation in rodents using a prevascularized site without graft support.

    Article  CAS  PubMed  Google Scholar 

  129. Gala-Lopez, B. et al. A novel pre-vascularized subcutaneous site safely accommodates stem cell derived therapies for treating diabetes. J. Stem Trans. Bio. 2, 107 (2016).

    Google Scholar 

  130. Gala-Lopez, B. L. et al. Subcutaneous clinical islet transplantation in a prevascularized subcutaneous pouch — preliminary experience. Cell R4 4, e2132 (2016).

    Google Scholar 

  131. O'Sullivan, E. S., Vegas, A., Anderson, D. G. & Weir, G. C. Islets transplanted in immunoisolation devices: a review of the progress and the challenges that remain. Endocr. Rev. 32, 827–844 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Agulnick, A. D. et al. Insulin-producing endocrine cells differentiated in vitro from human embryonic stem cells function in macroencapsulation devices in vivo. Stem Cells Transl Med. 4, 1214–1222 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Motte, E. et al. Composition and function of macroencapsulated human embryonic stem cell-derived implants: comparison with clinical human islet cell grafts. Am. J. Physiol. Endocrinol. Metab. 307, E838–E846 (2014).

    Article  CAS  PubMed  Google Scholar 

  134. Doloff, J. C. et al. Colony stimulating factor-1 receptor is a central component of the foreign body response to biomaterial implants in rodents and non-human primates. Nat. Mater. 16, 671–680 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Smith, K. E. et al. Acute ischemia induced by high density culture increases cytokine expression and diminishes the function and viability of highly purified human islets of langerhans. Transplantation http://dx.doi.org/10.1097/TP.0000000000001714 (2017).

  136. Uludag, H., De Vos, P. & Tresco, P. A. Technology of mammalian cell encapsulation. Adv. Drug Delivery Rev. 42, 29–64 (2000).

    Article  CAS  Google Scholar 

  137. Krishnan, R. et al. Noninvasive evaluation of the vascular response to transplantation of alginate encapsulated islets using the dorsal skin-fold model. Biomaterials 35, 891–898 (2014).

    Article  CAS  PubMed  Google Scholar 

  138. Chen, T. et al. Alginate encapsulant incorporating CXCL12 supports long-term allo- and xenoislet transplantation without systemic immune suppression. Am. J. Transplant. 15, 618–627 (2015).

    Article  CAS  PubMed  Google Scholar 

  139. Jacobs-Tulleneers-Thevissen, D. et al. Sustained function of alginate-encapsulated human islet cell implants in the peritoneal cavity of mice leading to a pilot study in a type 1 diabetic patient. Diabetologia 56, 1605–1614 (2013).

    Article  CAS  PubMed  Google Scholar 

  140. Dufrane, D., Goebbels, R. M., Saliez, A., Guiot, Y. & Gianello, P. Six-month survival of microencapsulated pig islets and alginate biocompatibility in primates: proof of concept. Transplantation 81, 1345–1353 (2006).

    Article  PubMed  Google Scholar 

  141. Garkavenko, O., Durbin, K., Tan, P. & Elliott, R. Islets transplantation: New Zealand experience. Xenotransplantation 18, 60 (2011).

    Article  Google Scholar 

  142. Basta, G. et al. Long-term metabolic and immunological follow-up of nonimmunosuppressed patients with type 1 diabetes treated with microencapsulated islet allografts. Diabetes Care 34, 2406–2409 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Tuch, B. E. et al. Safety and viability of microencapsulated human islets transplanted into diabetic humans. Diabetes Care 32, 1887–1889 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Vegas, A. J. et al. Long-term glycemic control using polymer-encapsulated human stem cell-derived beta cells in immune-competent mice. Nat. Med. 22, 306–311 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Mas, A. et al. Reversal of type 1 diabetes by engineering a glucose sensor in skeletal muscle. Diabetes 55, 1546–1553 (2006).

    Article  CAS  PubMed  Google Scholar 

  146. Lipes, M. A. et al. Insulin-secreting non-islet cells are resistant to autoimmune destruction. Proc. Natl Acad. Sci. USA 93, 8595–8600 (1996).

    Article  CAS  PubMed  Google Scholar 

  147. 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  Google Scholar 

  148. Xu, X. et al. Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell 132, 197–207 (2008). This work utilized the pancreatic duct ligation model to highlight the plasticity of the mouse pancreas, including the ability of NGN3 to induce the neogenesis of insulin-producing cells.

    Article  CAS  PubMed  Google Scholar 

  149. Swales, N. et al. Plasticity of adult human pancreatic duct cells by neurogenin3-mediated reprogramming. PLoS ONE 7, e37055 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  151. 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 

  152. Zhang, M. et al. Growth factors and medium hyperglycemia induce Sox9+ ductal cell differentiation into beta cells in mice with reversal of diabetes. Proc. Natl Acad. Sci. USA 113, 650–655 (2016).

    Article  CAS  PubMed  Google Scholar 

  153. Valdez, I. A. et al. Proinflammatory cytokines induce endocrine differentiation in pancreatic ductal cells via STAT3-dependent NGN3 activation. Cell Rep. 15, 460–470 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Cheng, C. et al. Fasting-mimicking diet promotes Ngn3-driven β-cell regeneration to reverse diabetes. Cell 168, 775–788 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Bonner-Weir, S. et al. In vitro cultivation of human islets from expanded ductal tissue. Proc. Natl Acad. Sci. USA 97, 7999–8004 (2000).

    Article  CAS  PubMed  Google Scholar 

  156. Lee, J. et al. Expansion and conversion of human pancreatic ductal cells into insulin-secreting endocrine cells. eLife 2, e00940 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Yamada, T. et al. Reprogramming mouse cells with a pancreatic duct phenotype to insulin-producing beta-like cells. Endocrinology 156, 2029–2038 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Gmyr, V. et al. Human pancreatic ductal cells: large-scale isolation and expansion. Cell Transplant. 10, 109–121 (2001).

    Article  CAS  PubMed  Google Scholar 

  159. Coad, R. A., Dutton, J. R., Tosh, D. & Slack, J. M. Inhibition of Hes1 activity in gall bladder epithelial cells promotes insulin expression and glucose responsiveness. Biochem. Cell Biol. 87, 975–987 (2009).

    Article  CAS  PubMed  Google Scholar 

  160. Hickey, R. D. et al. Generation of islet-like cells from mouse gall bladder by direct ex vivo reprogramming. Stem Cell Res. 11, 503–515 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Rahier, J., Wallon, J. & Henquin, J. C. Cell populations in the endocrine pancreas of human neonates and infants. Diabetologia 20, 540–546 (1981).

    Article  CAS  PubMed  Google Scholar 

  162. Rooman, I., Lardon, J., Flamez, D., Schuit, F. & Bouwens, L. Mitogenic effect of gastrin and expression of gastrin receptors in duct-like cells of rat pancreas. Gastroenterology 121, 940–949 (2001).

    Article  CAS  PubMed  Google Scholar 

  163. Rooman, I., Lardon, J. & Bouwens, L. Gastrin stimulates beta-cell neogenesis and increases islet mass from transdifferentiated but not from normal exocrine pancreas tissue. Diabetes 51, 686–690 (2002).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  165. Wang, T. C. et al. Pancreatic gastrin stimulates islet differentiation of transforming growth factor alpha-induced ductular precursor cells. J. Clin. Invest. 92, 1349–1356 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Rooman, I. & Bouwens, L. Combined gastrin and epidermal growth factor treatment induces islet regeneration and restores normoglycaemia in C57Bl6/J mice treated with alloxan. Diabetologia 47, 259–265 (2004).

    Article  CAS  PubMed  Google Scholar 

  167. Suarez-Pinzon, W. L., Lakey, J. R., Brand, S. J. & Rabinovitch, A. Combination therapy with epidermal growth factor and gastrin induces neogenesis of human islet β-cells from pancreatic duct cells and an increase in functional β-cell mass. J. Clin. Endocrinol. Metab. 90, 3401–3409 (2005).

    Article  CAS  PubMed  Google Scholar 

  168. Baeyens, L. et al. Transient cytokine treatment induces acinar cell reprogramming and regenerates functional beta cell mass in diabetic mice. Nat. Biotechnol. 32, 76–83 (2014). Here the authors demonstrated that, in states of chronic diabetes, the use of cytokines is sufficient for the regeneration of β cells from acinar tissue in mice.

    Article  CAS  PubMed  Google Scholar 

  169. Blaine, S. A. et al. Adult pancreatic acinar cells give rise to ducts but not endocrine cells in response to growth factor signaling. Development 137, 2289–2296 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Zhou, Q., Brown, J., Kanarek, A., Rajagopal, J. & Melton, D. A. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature 455, 627–632 (2008). This paper utilized a positive and negative selection scheme to identify three transcription factors that were able to induce the transdifferentiation of acinar tissue into insulin producing cells.

    Article  CAS  PubMed  Google Scholar 

  171. Li, W. et al. In vivo reprogramming of pancreatic acinar cells to three islet endocrine subtypes. eLife 3, e01846 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Li, W. et al. Long-term persistence and development of induced pancreatic beta cells generated by lineage conversion of acinar cells. Nat. Biotechnol. 32, 1223–1230 (2014).

    Article  CAS  PubMed  Google Scholar 

  173. Cavelti-Weder, C. et al. Hyperglycaemia attenuates in vivo reprogramming of pancreatic exocrine cells to beta cells in mice. Diabetologia 59, 522–532 (2016).

    Article  CAS  PubMed  Google Scholar 

  174. Clayton, H. W. et al. Pancreatic inflammation redirects acinar to beta cell reprogramming. Cell Rep. 17, 2028–2041 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Lima, M. J. et al. Generation of functional beta-like cells from human exocrine pancreas. PLoS ONE 11, e0156204 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Lima, M. J. et al. Suppression of epithelial-to-mesenchymal transitioning enhances ex vivo reprogramming of human exocrine pancreatic tissue toward functional insulin-producing beta-like cells. Diabetes 62, 2821–2833 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Park, J. Y. et al. Pdx1 expression in pancreatic precursor lesions and neoplasms. Appl. Immunohistochem. Mol. Morphol. 19, 444–449 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Meier, J. J. et al. Beta-cell replication is the primary mechanism subserving the postnatal expansion of beta-cell mass in humans. Diabetes 57, 1584–1594 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Van Assche, F. A., Aerts, L. & De Prins, F. A morphological study of the endocrine pancreas in human pregnancy. Br. J. Obstet. Gynaecol. 85, 818–820 (1978).

    Article  CAS  PubMed  Google Scholar 

  180. Saisho, Y. et al. beta-cell mass and turnover in humans: effects of obesity and aging. Diabetes Care 36, 111–117 (2013).

    Article  PubMed  Google Scholar 

  181. Fiaschi-Taesch, N. M. et al. Human pancreatic β-cell G1/S molecule cell cycle atlas. Diabetes 62, 2450–2459 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Shirikawa, Jun et al. Insulin signaling regulates the FoxM1/PLK1/CENP-A pathway to promote adaptive pancreatic β cell proliferation. Cell. Metabolism 25, 868–882.e5 (2017).

    Article  CAS  Google Scholar 

  183. Folli, F. et al. Altered insulin receptor signalling and beta-cell cycle dynamics in type 2 diabetes mellitus. PLoS ONE 6, e28050 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Kim, H. et al. Serotonin regulates pancreatic beta cell mass during pregnancy. Nat. Med. 16, 804–808 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Rieck, S. & Kaestner, K. H. Expansion of beta-cell mass in response to pregnancy. Trends Endocrinol. Metab. 21, 151–158 (2010).

    Article  CAS  PubMed  Google Scholar 

  186. Lakshmipathi, J. et al. PKCzeta is essential for pancreatic beta-cell replication during insulin resistance by regulating mTOR and Cyclin-D2. Diabetes 65, 1283–1296 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Fiaschi-Taesch, N. et al. Survey of the human pancreatic beta-cell G1/S proteome reveals a potential therapeutic role for cdk-6 and cyclin D1 in enhancing human beta-cell replication and function in vivo. Diabetes 58, 882–893 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Dirice, E. et al. Inhibition of DYRK1A stimulates human beta-cell proliferation. Diabetes 65, 1660–1671 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Shen, W. et al. Inhibition of DYRK1A and GSK3B induces human beta-cell proliferation. Nat. Commun. 6, 8372 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Wang, P. et al. A high-throughput chemical screen reveals that harmine-mediated inhibition of DYRK1A increases human pancreatic beta cell replication. Nat. Med. 21, 383–388 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. El Ouaamari, A. et al. SerpinB1 promotes pancreatic beta cell proliferation. Cell Metab. 23, 194–205 (2016).

    Article  CAS  PubMed  Google Scholar 

  192. Wang, P. et al. Diabetes mellitus — advances and challenges in human beta-cell proliferation. Nat. Rev. Endocrinol. 11, 201–212 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Chera, S. et al. Diabetes recovery by age-dependent conversion of pancreatic delta-cells into insulin producers. Nature 514, 503–507 (2014). This manuscript demonstrated that regeneration of insulin-producing cells occurs in unique ways depending on the age of the mouse: prior to puberty, δ cells are predominantly responsible for β-cell regeneration.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. van der Meulen, T. et al. Virgin beta cells persist throughout life at a neogenic niche within pancreatic islets. Cell Metab. 25, 911–926.e6 (2017).

    Article  CAS  PubMed  Google Scholar 

  196. Beamish, C. et al. Insulin-positive, Glut2-low cells present within mouse pancreas exhibit lineage plasticity and are enriched within extra-islet endocrine cell clusters. Islets 18, 65–82 (2016).

    Article  CAS  Google Scholar 

  197. Razavi, R. et al. Diabetes enhances the proliferation of adult pancreatic multipotent progenitor cells and biases their differentiation to more β-Cell production. Diabetes 64, 1311–1323 (2015).

    Article  CAS  PubMed  Google Scholar 

  198. Bader, E. et al. Identification of proliferative and mature β-cells in the islets of Langerhans. Nature 535, 430–434 (2016).

    Article  CAS  PubMed  Google Scholar 

  199. Collombat, P. et al. The ectopic expression of Pax4 in the mouse pancreas converts progenitor cells into alpha and subsequently beta cells. Cell 138, 449–462 (2009). Here the authors provide conclusive evidence that ectopic expression of Pax4 is sufficient for the transdifferentiation of α cells to β cells in mice.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Al-Hasani, K. et al. Adult duct-lining cells can reprogram into beta-like cells able to counter repeated cycles of toxin-induced diabetes. Dev. Cell 26, 86–100 (2013).

    Article  CAS  PubMed  Google Scholar 

  201. Collombat, P. et al. Opposing actions of Arx and Pax4 in endocrine pancreas development. Genes Dev. 17, 2591–2603 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Courtney, M. et al. The inactivation of Arx in pancreatic alpha-cells triggers their neogenesis and conversion into functional beta-like cells. PLoS Genet. 9, e1003934 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Chi, N. & Epstein, J. A. Getting your Pax straight: Pax proteins in development and disease. Trends Genet. 18, 41–47 (2002).

    Article  CAS  PubMed  Google Scholar 

  204. Ben-Othman, N. et al. Long-term GABA administration induces alpha cell-mediated beta-like cell neogenesis. Cell 168, 73–85.e11 (2016). This paper demonstrated that the neurotransmitter GABA can promote transdifferentiation of α cells to β cells in mice.

    Article  CAS  PubMed  Google Scholar 

  205. Li, J. et al. Artemisinins target GABAA receptor signaling and impair alpha cell identity. Cell 168, 86–100.e15 (2016). This paper identifies artemisinins as clinically relevant medicines that can target GABA receptors on α cells to induced their transdifferentiation into β cells in mice and isolated human islets.

    Article  CAS  PubMed  Google Scholar 

  206. Gonzalez, L. A., Gatch, M. B., Forster, M. J. & Dillon, G. H. Abuse potential of Soma: the GABA(A) receptor as a target. Mol. Cell. Pharmacol. 1, 180–186 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Longo, L. P. & Johnson, B. Addiction: Part, I. Benzodiazepines — side effects, abuse risk and alternatives. Am. Fam. Physician 61, 2121–2128 (2000).

    CAS  PubMed  Google Scholar 

  208. Slack, J. M. Developmental biology of the pancreas. Development 121, 1569–1580 (1995).

    CAS  PubMed  Google Scholar 

  209. Offield, M. F. et al. PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development 122, 983–995 (1996).

    CAS  PubMed  Google Scholar 

  210. Ferber, S. et al. Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia. Nat. Med. 6, 568–572 (2000).

    Article  CAS  PubMed  Google Scholar 

  211. Ber, I. et al. Functional, persistent, and extended liver to pancreas transdifferentiation. J. Biol. Chem. 278, 31950–31957 (2003).

    Article  CAS  PubMed  Google Scholar 

  212. 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  Google Scholar 

  213. Yechoor, V. et al. Neurogenin3 is sufficient for transdetermination of hepatic progenitor cells into neo-islets in vivo but not transdifferentiation of hepatocytes. Dev. Cell 16, 358–373 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Donelan, W. et al. Pancreatic and duodenal homeobox gene 1 (Pdx1) down-regulates hepatic transcription factor 1 alpha (HNF1alpha) expression during reprogramming of human hepatic cells into insulin-producing cells. Am. J. Transl Res. 7, 995–1008 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  215. Tang, D. Q. et al. Genetically reprogrammed, liver-derived insulin-producing cells are glucose-responsive, but susceptible to autoimmune destruction in settings of murine model of type 1 diabetes. Am. J. Transl Res. 5, 184–199 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  216. Banga, A., Akinci, E., Greder, L. V., Dutton, J. R. & Slack, J. M. In vivo reprogramming of Sox9+ cells in the liver to insulin-secreting ducts. Proc. Natl Acad. Sci. USA 109, 15336–15341 (2012).

    Article  PubMed  Google Scholar 

  217. Banga, A., Greder, L. V., Dutton, J. R. & Slack, J. M. Stable insulin-secreting ducts formed by reprogramming of cells in the liver using a three-gene cocktail and a PPAR agonist. Gene Ther. 21, 19–27 (2014).

    Article  CAS  PubMed  Google Scholar 

  218. Nagasaki, H. et al. Generation of insulin-producing cells from the mouse liver using beta cell-related gene transfer including Mafa and Mafb. PLoS ONE 9, e113022 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Ham, D. S. et al. Generation of functional insulin-producing cells from neonatal porcine liver-derived cells by PDX1/VP16, BETA2/NeuroD and MafA. PLoS ONE 8, e79076 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Li, R. et al. Gene therapy with neurogenin3, betacellulin and SOCS1 reverses diabetes in NOD mice. Gene Ther. 22, 876–882 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Di Stasi, A. et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N. Engl. J. Med. 365, 1673–1683 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Kolodka, T. M., Finegold, M., Moss, L. & Woo, S. L. Gene therapy for diabetes mellitus in rats by hepatic expression of insulin. Proc. Natl Acad. Sci. USA 92, 3293–3297 (1995). This paper demonstrated that hepatocytes can secrete insulin to provide therapy in diabetic rats.

    Article  CAS  PubMed  Google Scholar 

  223. Ren, B. et al. Long-term correction of diabetes in rats after lentiviral hepatic insulin gene therapy. Diabetologia 50, 1910–1920 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Elsner, M., Jorns, A. & Lenzen, S. Diabetes therapy by lentiviral hepatic insulin gene expression without transformation of liver. Diabetologia 51, 694–695 (2008).

    Article  CAS  PubMed  Google Scholar 

  225. Elsner, M. et al. Reversal of diabetes through gene therapy of diabetic rats by hepatic insulin expression via lentiviral transduction. Mol. Ther. 20, 918–926 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Hsu, P. Y., Kotin, R. M. & Yang, Y. W. Glucose- and metabolically regulated hepatic insulin gene therapy for diabetes. Pharm. Res. 25, 1460–1468 (2008).

    Article  CAS  PubMed  Google Scholar 

  227. Thule, P. M. et al. Long-term glycemic control with hepatic insulin gene therapy in streptozotocin-diabetic mice. J. Gene Med. 17, 141–152 (2015).

    Article  CAS  PubMed  Google Scholar 

  228. Thule, P. M. et al. Hepatic insulin gene therapy prevents deterioration of vascular function and improves adipocytokine profile in STZ-diabetic rats. Am. J. Physiol. Endocrinol. Metab. 290, E114–E122 (2006).

    Article  CAS  PubMed  Google Scholar 

  229. You, S. et al. Hepatic insulin gene therapy prevents diabetic enteropathy in STZ-treated CD-1 mice. Mol. Ther. Methods Clin. Dev. 2, 15028 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. You, S. et al. Effects of hepatic insulin gene therapy on enteric neuropathy in STZ-diabetic mice [Chinese]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 36, 546–553 (2011).

    CAS  PubMed  Google Scholar 

  231. Okitsu, T. et al. Transplantation of reversibly immortalized insulin-secreting human hepatocytes controls diabetes in pancreatectomized pigs. Diabetes 53, 105–112 (2004).

    Article  CAS  PubMed  Google Scholar 

  232. Olson, D. E., Paveglio, S. A., Huey, P. U., Porter, M. H. & Thule, P. M. Glucose-responsive hepatic insulin gene therapy of spontaneously diabetic BB/Wor rats. Hum. Gene Ther. 14, 1401–1413 (2003).

    Article  CAS  PubMed  Google Scholar 

  233. Groskreutz, D. J., Sliwkowski, M. X. & Gorman, C. M. Genetically engineered proinsulin constitutively processed and secreted as mature, active insulin. J. Biol. Chem. 269, 6241–6245 (1994).

    CAS  PubMed  Google Scholar 

  234. Thule, P. M. & Liu, J. M. Regulated hepatic insulin gene therapy of STZ-diabetic rats. Gene Ther. 7, 1744–1752 (2000).

    Article  CAS  PubMed  Google Scholar 

  235. Jensen, J. et al. Control of endodermal endocrine development by Hes-1. Nat. Genet. 24, 36–44 (2000).

    Article  CAS  PubMed  Google Scholar 

  236. Schonhoff, S. E., Giel-Moloney, M. & Leiter, A. B. Minireview: development and differentiation of gut endocrine cells. Endocrinology 145, 2639–2644 (2004).

    Article  CAS  PubMed  Google Scholar 

  237. Fujita, Y., Cheung, A. T. & Kieffer, T. J. Harnessing the gut to treat diabetes. Pediatr. Diabetes 5 (Suppl. 2), 57–69 (2004).

    Article  PubMed  Google Scholar 

  238. Kitamura, T. et al. Regulation of pancreatic juxtaductal endocrine cell formation by FoxO1. Mol. Cell. Biol. 29, 4417–4430 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Talchai, C., Xuan, S., Kitamura, T., DePinho, R. A. & Accili, D. Generation of functional insulin-producing cells in the gut by Foxo1 ablation. Nat. Genet. 44, 406–412, S401 (2012). In this manuscript, the authors demonstrated that suppression of FOXO1 could induce the generation of insulin-producing cells in the gut that could reverse toxin-induced diabetes in mice.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Chen, Y. J. et al. De novo formation of insulin-producing “neo-beta cell islets” from intestinal crypts. Cell Rep. 6, 1046–1058 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Ariyachet, C. et al. Reprogrammed stomach tissue as a renewable source of functional beta cells for blood glucose regulation. Cell Stem Cell 18, 410–421 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Young, L. S. & Mautner, V. The promise and potential hazards of adenovirus gene therapy. Gut 48, 733–736 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Kriegel, C., Attarwala, H. & Amiji, M. Multi-compartmental oral delivery systems for nucleic acid therapy in the gastrointestinal tract. Adv. Drug Deliv. Rev. 65, 891–901 (2013).

    Article  CAS  PubMed  Google Scholar 

  244. Wang, Y. et al. Conversion of human gastric epithelial cells to multipotent endodermal progenitors using defined small molecules. Cell Stem Cell 19, 449–461 (2016).

    Article  CAS  PubMed  Google Scholar 

  245. Isenman, L., Liebow, C. & Rothman, S. The endocrine secretion of mammalian digestive enzymes by exocrine glands. Am. J. Physiol. 276, E223–E232 (1999).

    CAS  PubMed  Google Scholar 

  246. Samuni, Y. & Baum, B. J. Gene delivery in salivary glands: from the bench to the clinic. Biochim. Biophys. Acta 1812, 1515–1521 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  247. Goldfine, I. D. et al. The endocrine secretion of human insulin and growth hormone by exocrine glands of the gastrointestinal tract. Nat. Biotechnol. 15, 1378–1382 (1997).

    Article  CAS  PubMed  Google Scholar 

  248. Rowzee, A. M. et al. Expression and secretion of human proinsulin-B10 from mouse salivary glands: implications for the treatment of type I diabetes mellitus. PLoS ONE 8, e59222 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Baum, B. J. et al. Early responses to adenoviral-mediated transfer of the aquaporin-1 cDNA for radiation-induced salivary hypofunction. Proc. Natl Acad. Sci. USA 109, 19403–19407 (2012).

    Article  PubMed  Google Scholar 

  250. Baum, B. J., Alevizos, I., Chiorini, J. A., Cotrim, A. P. & Zheng, C. Advances in salivary gland gene therapy — oral and systemic implications. Expert Opin. Biol. Ther. 15, 1443–1454 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Cheung, A. T. et al. Glucose-dependent insulin release from genetically engineered K cells. Science 290, 1959–1962 (2000). This manuscript demonstated the possibility of using genetic engineering to target glucose-reponsive cells in the gut for insulin replacement to treat diabetes.

    Article  CAS  PubMed  Google Scholar 

  252. Mojibian, M. et al. Insulin-producing intestinal K cells protect nonobese diabetic mice from autoimmune diabetes. Gastroenterology 147, 162–171.e6 (2014).

    Article  CAS  PubMed  Google Scholar 

  253. Mazzola, N. Review of current and emerging therapies in type 2 diabetes mellitus. Am. J. Manag. Care 18, S17–S26 (2012).

    PubMed  Google Scholar 

  254. Russell, S. J. et al. Outpatient glycemic control with a bionic pancreas in type 1 diabetes. N. Engl. J. Med. 371, 313–325 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  255. 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  PubMed  Google Scholar 

  256. International Diabetes Foundation. IDF Diabetes 7th edn (International Diabetes Foundation, 2015).

  257. Anderson, R. J., Freedland, K. E., Clouse, R. E. & Lustman, P. J. The prevalence of comorbid depression in adults with diabetes — a meta-analysis. Diabetes Care 24, 1069–1078 (2001).

    Article  CAS  PubMed  Google Scholar 

  258. American Diabetes Association. Economic costs of diabetes in the U.S. in 2012. Diabetes Care 36, 1033–1046 (2013).

  259. Banting, F. G., Best, C. H., Collip, J. B., Campbell, W. R. & Fletcher, A. A. Pancreatic extracts in the treatment of diabetes mellitus. Can. Med. Assoc. J. 7, 141–146 (1922). This classic paper details the exceedingly important discovery of insulin, for which Banting shared the 1923 Nobel Prize in Physiology or Medicine.

    Google Scholar 

  260. Rawshani, A., Svensson, A.-M., Rosengren, A., Eliasson, B. & Gudbjörnsdottir, S. Impact of socioeconomic status on cardiovascular disease and mortality in 24,947 individuals with type 1 diabetes. Diabetes Care 38, 1518–1527 (2015).

    Article  PubMed  Google Scholar 

  261. Fuchsberger, C. et al. The genetic architecture of type 2 diabetes. Nature 536, 41–47 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. Anik, A., Catli, G., Abaci, A. & Bober, E. Maturity-onset diabetes of the young (MODY): an update. J. Pediatr. Endocrinol. Metab. 28, 251–263 (2015).

    CAS  PubMed  Google Scholar 

  263. Williams, P. Notes on diabetes treated with extract and by grafts of sheep's pancreas. Br. Med. J. 2, 1303–1304 (1894).

    Google Scholar 

  264. 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). This publication describes the 'Edmonton Protocol' for effective treatment of T1DM by islet transplantation.

    Article  CAS  PubMed  Google Scholar 

  265. Lardon, J. & Bouwens, L. Metaplasia in the pancreas. Differentiation 73, 278–286 (2005).

    Article  CAS  PubMed  Google Scholar 

  266. Jemal, A., Siegel, R., Xu, J. & Ward, E. Cancer statistics, 2010. CA Cancer J. Clin. 60, 277–300 (2010).

    Article  PubMed  Google Scholar 

  267. Peery, A. F. et al. Burden of gastrointestinal disease in the United States: 2012 update. Gastroenterology 143, 1179–1187.e1-3 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  268. Sankaran, S. J. et al. Frequency of progression from acute to chronic pancreatitis and risk factors: a meta-analysis. Gastroenterology 149, 1490–1500.e1 (2015).

    Article  PubMed  Google Scholar 

  269. Whitcomb, D. C. et al. Common genetic variants in the CLDN2 and PRSS1-PRSS2 loci alter risk for alcohol-related and sporadic pancreatitis. Nat. Genet. 44, 1349–1354 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Pfützer, R. H. et al. SPINK1/PSTI polymorphisms act as disease modifiers in familial and idiopathic chronic pancreatitis. Gastroenterology 119, 615–623 (2000).

    Article  PubMed  Google Scholar 

  271. Kloppel, G. Chronic pancreatitis, pseudotumors and other tumor-like lesions. Mod. Pathol. 20, S113–S131 (2007).

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to the review.

Corresponding author

Correspondence to Timothy J. Kieffer.

Ethics declarations

Competing interests

T.J.K. has patents related to the material covered in this Review and is a co-founder and shareholder of enGene, a biotechnology company developing gene therapy approaches for metabolic diseases, including diabetes. He has also collaborated with scientists at BetaLogics and ViaCyte on stem-cell-based therapies for diabetes and received funding from Janssen R&D for this research.

Related links

PowerPoint slides

Glossary

Exocrine

Pertaining to the secretion of products from glands into ducts.

Endocrine

Pertaining to the secretion of hormones directly into the bloodstream.

Pancreatic insufficiency

A state of inadequate exocrine secretions by the pancreas leading to incomplete digestion of food.

Reprogramming

The directed conversion of one cell type to another artificially through the introduction of exogenous factors.

Islets

The endocrine micro-organs of the pancreas, containing cells that secrete insulin, glucagon, or other hormones.

Acinar cell

The primary exocrine cell of the pancreas that secretes digestive enzymes.

Regeneration

The renewal, regrowth or restoration of cells or tissues.

Shwachman–Diamond syndrome

A rare congenital disorder presenting with many abnormalities including pancreatic insufficiency.

Pancreatectomy

The surgical removal of the pancreas.

Ductal cells

Duct-lining cells that carry exocrine secretions, such as the digestive enzymes of the pancreas.

Xenotransplantation

A type of transplantation in which the recipient and donor tissues are from different species.

Endoderm

One of the three germ layers that mainly gives rise to the gastrointestinal tract, including the stomach, intestine, pancreas and liver.

Macroencapsulation

The enclosure and containment of a large quantity of cells or tissue in a device for implantation.

Transdifferentiation

The process in which a mature cell converts to another cell type without returning to a stem-cell state.

Omentum

A folded layer of peritoneum that hangs in front of the abdominal viscera.

Microencapsulation

The enclosure and containment of small cell clusters or dispersed cells for implantation.

Gene therapy

A therapeutic approach involving the delivery of genes into cells to repair, replace or alter expression of abnormal, insufficient or targeted genes.

Gastrin

A peptide hormone secreted predominantly by G cells of the stomach that stimulates the production of acid by parietal cells.

Cell differentiation

The process whereby a cell becomes specialized to perform a specific function.

K cells

An intestinal endocrine cell that secretes glucose-dependent insulinotropic polypeptide.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ellis, C., Ramzy, A. & Kieffer, T. Regenerative medicine and cell-based approaches to restore pancreatic function. Nat Rev Gastroenterol Hepatol 14, 612–628 (2017). https://doi.org/10.1038/nrgastro.2017.93

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrgastro.2017.93

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