Korean Diabetes J.  2010 Apr;34(2):77-83. 10.4093/kdj.2010.34.2.77.

Cell Replacement and Regeneration Therapy for Diabetes

Affiliations
  • 1Lee Gil Ya Cancer and Diabetes Institute, Gachon University of Medicine and Science, Incheon, Korea. hsjeon@gachon.ac.kr

Abstract

Reduction of beta cell function and a beta cell mass is observed in both type 1 and type 2 diabetes. Therefore, restoration of this deficiency might be a therapeutic option for treatment of diabetes. Islet transplantation has benefits, such as reduced incidence of hypoglycemia and achievement of insulin independence. However, the major drawback is an insufficient supply of islet donors. Transplantation of cells differentiated in vitro or in vivo regeneration of insulin-producing cells are possible approaches for beta cell/islet regenerative therapy. Embryonic and adult stem cells, pancreatic ductal progenitor cells, acinar cells, and other endocrine cells have been shown to differentiate into pancreatic beta cells. Formation of fully functional beta cells and the safety of these cells are critical issues for successful clinical application.

Keyword

Beta cell; Diabetes mellitus; Differentiation; Islets of Langerhans; Regeneration; Stem cells

MeSH Terms

Achievement
Acinar Cells
Adult Stem Cells
Diabetes Mellitus
Endocrine Cells
Humans
Hypoglycemia
Incidence
Insulin
Insulin-Secreting Cells
Islets of Langerhans
Islets of Langerhans Transplantation
Pancreatic Ducts
Regeneration
Stem Cells
Tissue Donors
Transplants
Insulin

Figure

  • Fig. 1 Possible strategies for beta cell replacement and regeneration therapy. Insulin-producing cells can be restored by transplantation of cells derived from in vitro or in vivo regeneration. Transplantation of islets from normal subjects, insulin-producing cells differentiated from stem and/or progenitor cells in vitro, or non-β cells (e.g., hepatocytes) engineered to produce insulin can result in insulin production and control blood glucose levels. Introduction of stem cells, β cell growth factors or stimulation of β cell differentiation transcription factors can regenerate β cells in vivo, which then produce insulin and control blood glucose levels.


Reference

1. Wajchenberg BL. Beta-cell failure in diabetes and preservation by clinical treatment. Endocr Rev. 2007. 28:187–218.
2. Rhodes CJ. Type 2 diabetes: a matter of beta-cell life and death? Science. 2005. 307:380–384.
3. Adorini L, Gregori S, Harrison LC. Understanding autoimmune diabetes: insights from mouse models. Trends Mol Med. 2002. 8:31–38.
4. Robertson RP, Holohan TV, Genuth S. Therapeutic controversy: pancreas transplantation for type I diabetes. J Clin Endocrinol Metab. 1998. 83:1868–1874.
5. Scharp DW, Lacy PE, Santiago JV, McCullough CS, Weide LG, Boyle PJ, Falqui L, Marchetti P, Ricordi C, Gingerich RL, Jaffe AS, Cryer PE, Hanto DW, Anderson CB, Flye MW. Results of our first nine intraportal islet allografts in type 1, insulin-dependent diabetic patients. Transplantation. 1991. 51:76–85.
6. Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, Kneteman NM, Rajotte RV. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000. 343:230–238.
7. Alejandro R, Barton FB, Hering BJ, Wease S. 2008 Update from the Collaborative Islet Transplant Registry. Transplantation. 2008. 86:1783–1788.
8. Jun HS, Yoon JW. Approaches for the cure of type 1 diabetes by cellular and gene therapy. Curr Gene Ther. 2005. 5:249–262.
9. de Groot M, Schuurs TA, van Schilfgaarde R. Causes of limited survival of microencapsulated pancreatic islet grafts. J Surg Res. 2004. 121:141–150.
10. Hering BJ, Wijkstrom M, Graham ML, Hardstedt M, Aasheim TC, Jie T, Ansite JD, Nakano M, Cheng J, Li W, Moran K, Christians U, Finnegan C, Mills CD, Sutherland DE, Bansal-Pakala P, Murtaugh MP, Kirchhof N, Schuurman HJ. Prolonged diabetes reversal after intraportal xenotransplantation of wild-type porcine islets in immunosuppressed nonhuman primates. Nat Med. 2006. 12:301–303.
11. Hecht G, Eventov-Friedman S, Rosen C, Shezen E, Tchorsh D, Aronovich A, Freud E, Golan H, El-Hasid R, Katchman H, Hering BJ, Zung A, Kra-Oz Z, Shaked-Mishan P, Yusim A, Shtabsky A, Idelevitch P, Tobar A, Harmelin A, Bachar-Lustig E, Reisner Y. Embryonic pig pancreatic tissue for the treatment of diabetes in a nonhuman primate model. Proc Natl Acad Sci U S A. 2009. 106:8659–8664.
12. Efrat S, Fusco-DeMane D, Lemberg H, al Emran O, Wang X. Conditional transformation of a pancreatic beta-cell line derived from transgenic mice expressing a tetracycline-regulated oncogene. Proc Natl Acad Sci U S A. 1995. 92:3576–3580.
13. Narushima M, Kobayashi N, Okitsu T, Tanaka Y, Li SA, Chen Y, Miki A, Tanaka K, Nakaji S, Takei K, Gutierrez AS, Rivas-Carrillo JD, Navarro-Alvarez N, Jun HS, Westerman KA, Noguchi H, Lakey JR, Leboulch P, Tanaka N, Yoon JW. A human beta-cell line for transplantation therapy to control type 1 diabetes. Nat Biotechnol. 2005. 23:1274–1282.
14. Chen NK, Wong JS, Kee IH, Lai SH, Thng CH, Ng WH, Ng RT, Tan SY, Lee SY, Tan ME, Sivalingam J, Chow PK, Kon OL. Nonvirally modified autologous primary hepatocytes correct diabetes and prevent target organ injury in a large preclinical model. PLoS One. 2008. 3:e1734.
15. Han D, Tian Y, Zhang M, Zhou Z, Lu J. Prevention and treatment of experimental autoimmune encephalomyelitis with recombinant adeno-associated virus-mediated alpha-melanocyte-stimulating hormone-transduced PLP139-151-specific T cells. Gene Ther. 2007. 14:383–395.
16. Chen NK, Tan SY, Udolph G, Kon OL. Insulin expressed from endogenously active glucose-responsive EGR1 promoter in bone marrow mesenchymal stromal cells as diabetes therapy. Gene Ther. Forthcoming 2010.
17. Raikwar SP, Zavazava N. Insulin producing cells derived from embryonic stem cells: are we there yet? J Cell Physiol. 2009. 218:256–263.
18. Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, Young H, Richardson M, Smart NG, Cunningham J, Agulnick AD, D'Amour KA, Carpenter MK, Baetge EE. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol. 2008. 26:443–452.
19. Jun HS, Park EY. Adult stem cells as a renewable source of insulin-producing cells. Int J Stem Cell. 2009. 2:115–121.
20. Xu X, D'Hoker J, Stange G, Bonne S, De Leu N, Xiao X, Van de Casteele M, Mellitzer G, Ling Z, Pipeleers D, Bouwens L, Scharfmann R, Gradwohl G, Heimberg H. Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell. 2008. 132:197–207.
21. Seaberg RM, Smukler SR, Kieffer TJ, Enikolopov G, Asghar Z, Wheeler MB, Korbutt G, van der Kooy D. Clonal identification of multipotent precursors from adult mouse pancreas that generate neural and pancreatic lineages. Nat Biotechnol. 2004. 22:1115–1124.
22. Kang HM, Kim J, Park S, Kim H, Kim KS, Lee EJ, Seo SI, Kang SG, Lee JE, Lim H. Insulin-secreting cells from human eyelid-derived stem cells alleviate type I diabetes in immunocompetent mice. Stem Cells. 2009. 27:1999–2008.
23. Lin G, Wang G, Liu G, Yang LJ, Chang LJ, Lue TF, Lin CS. Treatment of type 1 diabetes with adipose tissue-derived stem cells expressing pancreatic duodenal homeobox 1. Stem Cells Dev. 2009. 18:1399–1406.
24. Jun HS. In vivo regeneration of insulin-producing beta-cells. Adv Exp Med Biol. 2010. 654:627–640.
25. Suarez-Pinzon WL, Power RF, Yan Y, Wasserfall C, Atkinson M, Rabinovitch A. Combination therapy with glucagon-like peptide-1 and gastrin restores normoglycemia in diabetic NOD mice. Diabetes. 2008. 57:3281–3288.
26. Suarez-Pinzon WL, Yan Y, Power R, Brand SJ, Rabinovitch A. Combination therapy with epidermal growth factor and gastrin increases beta-cell mass and reverses hyperglycemia in diabetic NOD mice. Diabetes. 2005. 54:2596–2601.
27. Suarez-Pinzon WL, Lakey JR, Rabinovitch A. Combination therapy with glucagon-like peptide-1 and gastrin induces beta-cell neogenesis from pancreatic duct cells in human islets transplanted in immunodeficient diabetic mice. Cell Transplant. 2008. 17:631–640.
28. von Herrath M. E1-INT (Transition Therapeutics/Novo Nordisk). Curr Opin Investig Drugs. 2005. 6:1037–1042.
29. Fleming A, Rosenberg L. Prospects and challenges for islet regeneration as a treatment for diabetes: a review of islet neogenesis associated protein. J Diabetes Sci Technol. 2007. 1:231–244.
30. Taniguchi H, Yamato E, Tashiro F, Ikegami H, Ogihara T, Miyazaki J. Beta-cell neogenesis induced by adenovirus-mediated gene delivery of transcription factor pdx-1 into mouse pancreas. Gene Ther. 2003. 10:15–23.
31. Sun YX, Zhang XZ, Cheng H, Cheng SX, Zhuo RX. A low-toxic and efficient gene vector: carboxymethyl dextran-graft-polyethylenimine. J Biomed Mater Res A. 2008. 84:1102–1110.
32. Koya V, Lu S, Sun YP, Purich DL, Atkinson MA, Li SW, Yang LJ. Reversal of streptozotocin-induced diabetes in mice by cellular transduction with recombinant pancreatic transcription factor pancreatic duodenal homeobox-1: a novel protein transduction domain-based therapy. Diabetes. 2008. 57:757–769.
33. Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature. 2008. 455:627–632.
34. Ferber S, Halkin A, Cohen H, Ber I, Einav Y, Goldberg I, Barshack I, Seijffers R, Kopolovic J, Kaiser N, Karasik A. Pancreatic and duodenal homeobox gene 1 induces expression of insulin genes in liver and ameliorates streptozotocin-induced hyperglycemia. Nat Med. 2000. 6:568–572.
35. Kojima H, Fujimiya M, Matsumura K, Younan P, Imaeda H, Maeda M, Chan L. NeuroD-betacellulin gene therapy induces islet neogenesis in the liver and reverses diabetes in mice. Nat Med. 2003. 9:596–603.
36. Kaneto H, Nakatani Y, Miyatsuka T, Matsuoka TA, Matsuhisa M, Hori M, Yamasaki Y. PDX-1/VP16 fusion protein, together with NeuroD or Ngn3, markedly induces insulin gene transcription and ameliorates glucose tolerance. Diabetes. 2005. 54:1009–1022.
37. Yechoor V, Liu V, Espiritu C, Paul A, Oka K, Kojima H, Chan L. Neurogenin3 is sufficient for transdetermination of hepatic progenitor cells into neo-islets in vivo but not transdifferentiation of hepatocytes. Dev Cell. 2009. 16:358–373.
38. Koizumi M, Nagai K, Kida A, Kami K, Ito D, Fujimoto K, Kawaguchi Y, Doi R. Forced expression of PDX-1 induces insulin production in intestinal epithelia. Surgery. 2006. 140:273–280.
39. Nomura S, Nakamura T, Hashimoto T, Nishio Y, Maegawa H, Kudo M, Kashiwagi A. MafA differentiates rat intestinal cells into insulin-producing cells. Biochem Biophys Res Commun. 2006. 349:136–143.
40. Liu MJ, Han J, Lee YS, Park MS, Shin S, Jun HS. Amelioration of hyperglycemia by intestinal overexpression of glucagon-like peptide-1 in mice. J Mol Med. 2010. 88:351–358.
41. Kodama S, Kuhtreiber W, Fujimura S, Dale EA, Faustman DL. Islet regeneration during the reversal of autoimmune diabetes in NOD mice. Science. 2003. 302:1223–1227.
42. Voltarelli JC, Couri CE, Stracieri AB, Oliveira MC, Moraes DA, Pieroni F, Coutinho M, Malmegrim KC, Foss-Freitas MC, Simoes BP, Foss MC, Squiers E, Burt RK. Autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus. JAMA. 2007. 297:1568–1576.
Full Text Links
  • KDJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr