Skip to main content

Advertisement

Log in

Bioprinting an Artificial Pancreas for Type 1 Diabetes

  • Immunology, Transplantation, and Regenerative Medicine (L Piemonti and V Sordi, Section Editors)
  • Published:
Current Diabetes Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

Pancreatic islet cell transplantation is currently the only curative cell therapy for type 1 diabetes mellitus. However, its potential to treat many more patients is limited by several challenges. The emergence of 3D bioprinting technology from recent advances in 3D printing, biomaterials, and cell biology has provided the means to overcome these challenges.

Recent Findings

3D bioprinting allows for the precise fabrication of complex 3D architectures containing spatially distributed cells, biomaterials (bioink), and bioactive factors. Different strategies to capitalize on this ability have been investigated for the 3D bioprinting of pancreatic islets. In particular, with co-axial bioprinting technology, the co-printability of islets with supporting cells such as endothelial progenitor cells and regulatory T cells, which have been shown to accelerate revascularization of islets and improve the outcome of various transplantations, respectively, has been achieved.

Summary

3D bioprinting of islets for generation of an artificial pancreas is a newly emerging field of study with a vast potential to improve islet transplantation.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Gregory JM, Moore DJ. Type 1 diabetes mellitus. Pediatr Rev 2013. https://doi.org/10.1016/B978-1-4377-1604-7.00561-3.

    Chapter  Google Scholar 

  2. Forouhi NG, Wareham NJ. Epidemiology of diabetes. Medicine (Baltimore). 2014;42:698–702. https://doi.org/10.1383/medc.2006.34.2.57.

    Article  Google Scholar 

  3. Diabetes DOF. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2013;36:67–74. https://doi.org/10.2337/dc13-S067.

    Article  Google Scholar 

  4. Pathiraja V, Kuehlich JP, Campbell PD, Krishnamurthy B, Loudovaris T, Coates PTH, et al. Proinsulin-specific, HLA-DQ8, and HLA-DQ8-transdimer-restricted CD4+ T cells infiltrate islets in type 1 diabetes. Diabetes. 2015;64:172–82. https://doi.org/10.2337/db14-0858.

    Article  CAS  PubMed  Google Scholar 

  5. Rezania A, Bruin JE, Arora P, Rubin A, Batushansky I, Asadi A, et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol. 2014;32:1121–33. https://doi.org/10.1038/nbt.3033.

    Article  CAS  PubMed  Google Scholar 

  6. Prabakar KR, Domínguez-Bendala J, Damaris Molano R, Pileggi A, Villate S, Ricordi C, et al. Generation of glucose-responsive, insulin-producing cells from human umbilical cord blood-derived mesenchymal stem cells. Cell Transplant. 2012;21:1321–39. https://doi.org/10.3727/096368911X612530.

    Article  PubMed  Google Scholar 

  7. Agarwal A, Brayman KL. Update on islet cell transplantation for type 1 diabetes. Semin Interv Radiol. 2012;29:90–8. https://doi.org/10.1055/s-0032-1312569.

    Article  Google Scholar 

  8. Robertson RP, Davis C, Larsen J, Stratta R, Sutherland DER, American Diabetes Association. Pancreas and islet transplantation in type 1 diabetes. Diabetes Care. 2006;29:935. https://doi.org/10.2337/diacare.29.04.06.dc06-9908.

    Article  PubMed  Google Scholar 

  9. Chan C, Chim TM, Leung K, Tong C, Wong T, Leung GK. Simultaneous pancreas and kidney transplantation as the standard surgical treatment for diabetes mellitus patients with end-stage renal disease. Hong Kong Med J. 2016;22(1):62–9. https://doi.org/10.12809/hkmj154613.

    Article  CAS  PubMed  Google Scholar 

  10. Troppmann C. Complications after pancreas transplantation. Curr Opin Organ Transpl. 2010;15:112–8. https://doi.org/10.1097/MOT.0b013e3283355349.

    Article  Google Scholar 

  11. Vardanyan M, Parkin E, Gruessner C, Rilo HLR. Pancreas vs. islet transplantation: a call on the future. Curr Opin Organ Transplant. 2010;15:124–30. https://doi.org/10.1097/MOT.0b013e32833553f8.

    Article  PubMed  Google Scholar 

  12. Robertson RP. Islet transplantation as a treatment for diabetes — a work in progress. N Engl J Med. 2004;350:694–705. https://doi.org/10.1056/NEJMra032425.

    Article  CAS  PubMed  Google Scholar 

  13. Connell PJO, Holmes-Walker DJ, Goodman D, Hawthorne WJ, Loudovaris T, Gunton JE, et al. Multicenter Australian trial of islet transplantation: improving accessibility and outcomes. Am J Transplant. 2013;13:1850–8. https://doi.org/10.1002/ajt.12250.

    Article  Google Scholar 

  14. Ichii H, Ricordi C. Current status of islet cell transplantation. J Hepato-Biliary-Pancreat Surg. 2013;16:101–12. https://doi.org/10.1007/s00534-008-0021-2.Current.

    Article  Google Scholar 

  15. Lacy PE, Walker MM, Fink CJ, Louis S. Perifusion of isolated rat islets in vitro participation of the microtubular system in the biphasic release of insulin. Diabetes. 1972;21:987–98. https://doi.org/10.2337/diab.21.10.987.

    Article  CAS  PubMed  Google Scholar 

  16. Najarian JS, Sutherland DER, Baumgartner D, Burke B, Rynasiewicz JJ, Matas AJ, et al. Total or near total pancreatectomy and islet autotransplantation for treatment of chronic pancreatitis.pdf. Ann Surg. 1980;192:526–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Robertson RP, Lanz KJ, Sutherland DER, Kendall DM. Prevention of diabetes for up to 13 years by autoislet transplantation after pancreatectomy for chronic pancreatitis. Diabetes. 2001;50:48–50. https://doi.org/10.2337/diabetes.50.1.47.

    Article  Google Scholar 

  18. Shapiro AMJ, Lakey JRT, Ryan EA, Korbutt GS, Toth E, Warnock GL, et al. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocoricoid-free imunosuppressive regimen. N Engl J Med. 2000;343:230–8. https://doi.org/10.1056/NEJM200007273430401.

    Article  CAS  PubMed  Google Scholar 

  19. Ryan EA, Paty BW, Senior PA, Bigam D, Alfadhli E, Kneteman NM, et al. Five-year follow-up after clinical islet transplantation. Diabetes. 2005;54:2060–9.

    Article  CAS  PubMed  Google Scholar 

  20. Leitão CB, Cure P, Tharvanji T, Baidal DA, Alejandro R. Current challenges in islet transplantation. Curr Diab Rep. 2008;8:324–31. https://doi.org/10.1007/s11892-008-0057-3.

    Article  PubMed  Google Scholar 

  21. Rheinheimer J, Bauer AC, Silveiro SP, Estivalet AAF, Bouças AP, Rosa AR, et al. Human pancreatic islet transplantation: an update and description of the establishment of a pancreatic islet isolation laboratory. Arch Endocrinol Metab. 2015;59:161–70. https://doi.org/10.1590/2359-3997000000030.

    Article  PubMed  Google Scholar 

  22. Lacy PE, Kostianovsky M. Method for the isolation of intact islets of Langerhans from the rat pancreas. Diabetes. 1967;16:35–9. https://doi.org/10.2337/diab.16.1.35.

    Article  CAS  PubMed  Google Scholar 

  23. Carlsson PO, Palm F, Andersson A, Liss P. Markedly decreased oxygen tension in transplanted rat pancreatic islets irrespective of the implantation site. Diabetes. 2001;50:489–95. https://doi.org/10.2337/diabetes.50.3.489.

    Article  CAS  PubMed  Google Scholar 

  24. Bennet W, Sundberg B, Groth CG, Brendel MD, Brandhorst D, Brandhorst H, et al. Incompatibility between human blood and isolated islets of Langerhans: a finding with implications for clinical intraportal islet transplantation? Diabetes. 1999;48:1907–14. https://doi.org/10.2337/diabetes.48.10.1907.

    Article  CAS  PubMed  Google Scholar 

  25. Moberg L, Johansson H, Lukinius A, Berne C, Foss A, Källen R, et al. Production of tissue factor by pancreatic islet cells as a trigger of detrimental thrombotic reactions in clinical islet transplantation. Lancet. 2002;360:2039–45. https://doi.org/10.1016/S0140-6736(02)12020-4.

    Article  CAS  PubMed  Google Scholar 

  26. Van Der Windt DJ, Bottino R, Casu A, Campanile N, Cooper DKC. Rapid loss of intraportally transplanted islets: an overview of pathophysiology and preventive strategies. Xenotransplantation. 2007;14:288–97. https://doi.org/10.1111/j.1399-3089.2007.00419.x.

    Article  PubMed  Google Scholar 

  27. Cantarelli E, Piemonti L. Alternative transplantation sites for pancreatic islet grafts. Curr Diab Rep. 2011;11:364–74. https://doi.org/10.1007/s11892-011-0216-9.

    Article  PubMed  Google Scholar 

  28. Bennet W, Groth CG, 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. 2000;105:125–33. https://doi.org/10.1517/03009734000000059.

    Article  CAS  PubMed  Google Scholar 

  29. Margolis RN, Holup JJ, Selawry HP. Effects of intratesticular islet transplantation on hepatic glycogen metabolism in the rat. Diabetes Res Clin Pract. 1986;2:291–9. https://doi.org/10.1016/S0168-8227(86)80006-7.

    Article  CAS  PubMed  Google Scholar 

  30. Ar’Rajab A, Dawidson IJ, Harris RB, Sentementes JT. Immune privilege of the testis for islet xenotransplantation (rat to mouse). Cell Transplant. 2017;3:493–8. https://doi.org/10.1177/096368979400300606.

    Article  Google Scholar 

  31. Kemp CB, Knight MJ, Scharp DW, Ballinger WF, Lacy PE. Effect of transplantation site on the results of pancreatic islet isografts in diabetic rats. Diabetologia. 1973;9:486–91. https://doi.org/10.1007/BF00461694.

    Article  CAS  PubMed  Google Scholar 

  32. Melzi R, Sanvito F, Mercalli A, Andralojc K, Bonifacio E, Piemonti L. Intrahepatic islet transplant in the mouse: functional and morphological characterization. Cell Transplant. 2008;17:1361–70. https://doi.org/10.3727/096368908787648146.

    Article  CAS  PubMed  Google Scholar 

  33. Mao G, Chen G, Bai H, Song T, Wang Y. The reversal of hyperglycaemia in diabetic mice using PLGA scaffolds seeded with islet-like cells derived from human embryonic stem cells. Biomaterials. 2009;30:1706–14. https://doi.org/10.1016/j.biomaterials.2008.12.030.

    Article  CAS  PubMed  Google Scholar 

  34. De Vos P, Hamel AF, Tatarkiewicz K. Considerations for successful transplantation of encapsulated pancreatic islets. Diabetologia. 2002;45:159–73. https://doi.org/10.1007/s00125-001-0729-x.

    Article  PubMed  Google Scholar 

  35. Mallett AG, Korbutt GS. Alginate modification improves long-term survival and function of transplanted encapsulated islets. Tissue Eng Part A. 2009;15:1301–9. https://doi.org/10.1089/ten.tea.2008.0118.

    Article  CAS  PubMed  Google Scholar 

  36. Desai T, Shea LD. Advances in islet encapsulation technologies. Nat Rev Drug Discov. 2017;16:338–50. https://doi.org/10.1038/nrd.2016.232.

    Article  CAS  PubMed  Google Scholar 

  37. Vériter S, Mergen J, Goebbels R-M, Aouassar N, Grégoire C, Jordan B, et al. In vivo selection of biocompatible alginates for islet encapsulation and subcutaneous transplantation. Tissue Eng Part A. 2010;16:1503–13. https://doi.org/10.1089/ten.TEA.2009.0286.

    Article  PubMed  Google Scholar 

  38. Lum ZP, Tai IT, Krestow M, Norton J, Vacek I, Sun AM. Prolonged reversal of diabetic state in NOD mice by xenografts of microencapsulated rat islets. Diabetes. 1991;40:1511–6. https://doi.org/10.2337/diab.40.11.1511.

    Article  CAS  PubMed  Google Scholar 

  39. Cui W, Barr G, Faucher KM, Sun X-L, Safley SA, Weber CJ, et al. A membrane-mimetic barrier for islet encapsulation. Transplant Proc. 2004;36:1206–8. https://doi.org/10.1016/j.transproceed.2004.04.059.

    Article  CAS  PubMed  Google Scholar 

  40. Lamb M, Storrs R, Li S, Liang O, Laugenour K, Dorian R, et al. Function and viability of human islets encapsulated in alginate sheets: in vitro and in vivo culture. Transplant Proc. 2011;43:3265–6. https://doi.org/10.1016/j.transproceed.2011.10.028.

    Article  CAS  PubMed  Google Scholar 

  41. Zhi ZL, Kerby A, King AJF, Jones PM, Pickup JC. Nano-scale encapsulation enhances allograft survival and function of islets transplanted in a mouse model of diabetes. Diabetologia. 2012;55:1081–90. https://doi.org/10.1007/s00125-011-2431-y.

    Article  CAS  PubMed  Google Scholar 

  42. Ma M, Chiu A, Sahay G, Doloff JC, Dholakia N, Thakrar R, et al. Core-shell hydrogel microcapsules for improved islets encapsulation. Adv Healthc Mater. 2013;2:667–72. https://doi.org/10.1002/adhm.201200341.

    Article  CAS  PubMed  Google Scholar 

  43. Hobbs HA, Kendall WF, Darrabie M, Opara EC. Prevention of morphological changes in alginate microcapsules for islet xenotransplantation. J Investig Med. 2001;49:572–5. https://doi.org/10.2310/6650.2001.33722.

    Article  CAS  PubMed  Google Scholar 

  44. De Vos P, Hillebrands JL, De Haan BJ, Strubbe JH, Van Schilfgaarde R. Efficacy of a prevascularized expanded polytetrafluoroethylene solid support system as a transplantation site for pancreatic islets. Transplantation. 1997;63:824–30. https://doi.org/10.1097/00007890-199703270-00006.

    Article  PubMed  Google Scholar 

  45. Dufour JM, Rajotte RV, Zimmerman M, Rezania A, Kin T, Dixon DE, et al. Development of an ectopic site for islet transplantation, using biodegradable scaffolds. Tissue Eng. 2005;11:1323–31. https://doi.org/10.1089/ten.2005.11.1323.

    Article  CAS  PubMed  Google Scholar 

  46. Wang RN, Rosenberg L. Maintenance of beta-cell function and survival following islet isolation requires re-establishment of the islet-matrix relationship. J Endocrinol. 1999;163:181–90. https://doi.org/10.1677/joe.0.1630181.

    Article  CAS  PubMed  Google Scholar 

  47. Gibly RF, Zhang X, Graham ML, Hering BJ, Kaufman DB, Lowe WL, et al. Extrahepatic islet transplantation with microporous polymer scaffolds in syngeneic mouse and allogeneic porcine models. Biomaterials. 2011;32:9677–84. https://doi.org/10.1016/j.biomaterials.2011.08.084.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Berman DM, O’Neil JJ, Coffey LCK, Chaffanjon PCJ, Kenyon NM, Ruiz P, et al. Long-term survival of nonhuman primate islets implanted in an omental pouch on a biodegradable scaffold. Am J Transplant. 2009;9:91–104. https://doi.org/10.1111/j.1600-6143.2008.02489.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Blomeier H, Zhang X, Rives C, Brissova M, Hughes E, Baker M, et al. Polymer scaffolds as synthetic microenvironments for extrahepatic islet transplantation. Transplantation. 2006;82:452–9. https://doi.org/10.1097/01.tp.0000231708.19937.21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nat Biotechnol. 2014;32:773–85. https://doi.org/10.1038/nbt.2958.

    Article  CAS  PubMed  Google Scholar 

  51. Schubert C, van Langeveld MC, Donoso LA. Innovations in 3D printing: a 3D overview from optics to organs. Br J Ophthalmol. 2014;98:159–61. https://doi.org/10.1136/bjophthalmol-2013-304446.

    Article  PubMed  Google Scholar 

  52. Ozbolat IT, Chen H, Yu Y. Development of ‘multi-arm bioprinter’ for hybrid biofabrication of tissue engineering constructs. Robot Comput Integr Manuf. 2014;30:295–304. https://doi.org/10.1016/j.rcim.2013.10.005.

    Article  Google Scholar 

  53. Kolesky DB, Truby RL, Gladman AS, Busbee TA, Homan KA, Lewis JA. 3D bioprinting of vascularized, heterogeneous cell-laden tissue constructs. Adv Mater. 2014;26:3124–30. https://doi.org/10.1002/adma.201305506.

    Article  CAS  PubMed  Google Scholar 

  54. Poldervaart MT, Gremmels H, van Deventer K, Fledderus JO, Oner FC, Verhaar MC, et al. Prolonged presence of VEGF promotes vascularization in 3D bioprinted scaffolds with defined architecture. J Control Release. 2014;184:58–66. https://doi.org/10.1016/j.jconrel.2014.04.007.

    Article  CAS  PubMed  Google Scholar 

  55. Yue Z, Liu X, Coates PT, Wallace GG. Advances in printing biomaterials and living cells. Curr Opin Organ Transplant. 2016;21:467–75. https://doi.org/10.1097/MOT.0000000000000346.

    Article  CAS  PubMed  Google Scholar 

  56. Di Bella C, Duchi S, O’Connell CD, Blanchard R, Augustine C, Yue Z, et al. In situ handheld three-dimensional bioprinting for cartilage regeneration. J Tissue Eng Regen Med. 2018;12:611–21. https://doi.org/10.1002/term.2476.

    Article  CAS  PubMed  Google Scholar 

  57. Kesti M, Eberhardt C, Pagliccia G, Kenkel D, Grande D, Boss A, et al. Bioprinting complex cartilaginous structures with clinically compliant biomaterials. Adv Funct Mater. 2015;25:7406–17. https://doi.org/10.1002/adfm.201503423.

    Article  Google Scholar 

  58. Lee JW, Choi Y-J, Yong W-J, Pati F, Shim J-H, Kang KS, et al. Development of a 3D cell printed construct considering angiogenesis for liver tissue engineering. Biofabrication. 2016;8:015007. https://doi.org/10.1088/1758-5090/7/2/025009.

    Article  CAS  PubMed  Google Scholar 

  59. Gu Q, Tomaskovic-crook E, Lozano R, Chen Y, Kapsa RM, Zhou Q, et al. Functional 3D neural mini-tissues from printed gel-based bioink and human neural stem cells. Adv Healthc Mater. 2016;5:1429–38. https://doi.org/10.1002/adhm.201600095.

    Article  CAS  PubMed  Google Scholar 

  60. Derakhshanfar S, Mbeleck R, Xu K, Zhang X, Zhong W, Xing M. 3D bioprinting for biomedical devices and tissue engineering: a review of recent trends and advances. Bioact Mater Elsevier Ltd. 2018;3:144–56. https://doi.org/10.1016/j.bioactmat.2017.11.008.

    Article  Google Scholar 

  61. •• Marchioli G, Van Gurp L, Van Krieken PP, Stamatialis D, Engelse M, Van Blitterswijk CA, et al. Fabrication of three-dimensional bioplotted hydrogel scaffolds for islets of Langerhans transplantation. Biofabrication. 2015;7:025009. https://doi.org/10.1088/1758-5090/7/2/025009 3D bioprinting technology was used to print pancreatic islets using alginate-based bioinks. 3D bioprinted human islets were highly viable and morphologically sound.

    Article  CAS  PubMed  Google Scholar 

  62. •• Duin S, Schütz K, Ahlfeld T, Lehmann S, Lode A, Ludwig B, et al. 3D bioprinting of functional islets of Langerhans in an alginate/methylcellulose hydrogel blend. Adv Healthc Mater. 2019;1801631:1–14. https://doi.org/10.1002/adhm.201801631 3D bioprinting technology was used to print rat islets using an alginate/methylcellulose bioink with minimal impact on islet viability , morphology, and function.

    Article  CAS  Google Scholar 

  63. Penko D, Mohanasundaram D, Sen S, Mee C, Bonder CS, Coates PTH, et al. Incorporation of endothelial progenitor cells into mosaic pseudoislets. Islets. 2011;3:73–9. https://doi.org/10.4161/isl.3.3.15392.

    Article  PubMed  Google Scholar 

  64. •• Liu X, Carter SSD, Renes MJ, Kim J, Rojas-Canales DM, Penko D, et al. Development of a coaxial 3D printing platform for biofabrication of implantable islet-containing constructs. Adv Healthc Mater. 2019;8(7):e1801181. https://doi.org/10.1002/adhm.201801181 A co-axial bioprinter was developed in-house and alginate-gelatin methacryloyl bioink was formulated, for co-printing of islets with supporting cells such as endothelial progenitor cells and regulatory T cells. Co-printed mouse islets and endothelial progenitor cells were highly viable.

    Article  CAS  PubMed  Google Scholar 

  65. Lifson N, Lassa CV, Dixit PK. Relation between blood flow and morphology in islet organ of rat pancreas. Am J Phys. 1985;249:E43–8. https://doi.org/10.1152/ajpendo.1985.249.1.E43.

    Article  CAS  Google Scholar 

  66. Johansson A, Lau J, Sandberg M, Borg LAH, Magnusson PU, Carlsson P-O. Endothelial cell signalling supports pancreatic beta cell function in the rat. Diabetologia. 2009;52:2385–94. https://doi.org/10.1007/s00125-009-1485-6.

    Article  CAS  PubMed  Google Scholar 

  67. Davalli AM, Scaglia L, Zangen DH, Hollister J, Bonner-Weir S, Weir GC. Vulnerability of islets in the immediate posttransplantation period. Dynamic changes in structure and function. Diabetes. 1996;45:1161–7. https://doi.org/10.2337/diabetes.45.9.1161.

    Article  CAS  PubMed  Google Scholar 

  68. de Groot M, Schuurs T, Fekken S, Leuvenink H, van Schilfgaarde R, Keizer J. Response of encapsulated rat pancreatic islets to hypoxia. Cell Transplant. 2003;12:867–75. https://doi.org/10.3727/000000003771000219.

    Article  PubMed  Google Scholar 

  69. Campbell PD, Weinberg A, Chee J, Mariana L, Ayala R, Hawthorne WJ, et al. Expression of pro- and antiapoptotic molecules of the Bcl-2 family in human islets postisolation. Cell Transplant. 2012;21:49–60. https://doi.org/10.3727/096368911X566262.

    Article  PubMed  Google Scholar 

  70. Cantley J, Walters SN, Jung M, Weinberg A, Cowley MJ, Whitworth PT, et al. A preexistent hypoxic gene signature predicts impaired islet graft function and glucose homeostasis. Cell Transplant. 2013;22:2147–59. https://doi.org/10.3727/096368912X658728.

    Article  PubMed  Google Scholar 

  71. Vajkoczy P, Menger MD, Simpson E, Messmer K. Angiogenesis and vascularization of murine pancreatic islet isografts. Transplantation. 1995;60:123–7. https://doi.org/10.1097/00007890-199507000-00002.

    Article  CAS  PubMed  Google Scholar 

  72. Mattsson G, Jansson L, Carlsson P-O. Decreased vascular density in mouse pancreatic islets after transplantation. Diabetes. 2002;51:1362–6. https://doi.org/10.2337/diabetes.51.5.1362.

    Article  CAS  PubMed  Google Scholar 

  73. Cho H-J, Lee N, Lee JY, Choi YJ, Ii M, Wecker A, et al. Role of host tissues for sustained humoral effects after endothelial progenitor cell transplantation into the ischemic heart. J Exp Med. 2007;204:3257–69. https://doi.org/10.1084/jem.20070166.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med. 2003;9:702–12. https://doi.org/10.1038/nm0603-702.

    Article  CAS  PubMed  Google Scholar 

  75. Peiris HS, Bonder CS, Coates PT, Keating DJ, Jessup CF. The b-cell/EC axis: how do islet cells talk to each other? Diabetes Care. 2014;63:3–11. https://doi.org/10.2337/db13-0617.

    Article  CAS  Google Scholar 

  76. Kang S, Park HS, Jo A, Hong SH, Lee HN, Lee YY, et al. Endothelial progenitor cell cotransplantation enhances islet engraftment by rapid revascularization. Diabetes. 2012;61:866–76. https://doi.org/10.2337/db10-1492.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Quaranta P, Antonini S, Spiga S, Mazzanti B, Curcio M, Mulas G, et al. Co-transplantation of endothelial progenitor cells and pancreatic islets to induce long-lasting normoglycemia in streptozotocin-treated diabetic rats. PLoS One. 2014;9:1–13. https://doi.org/10.1371/journal.pone.0094783.

    Article  CAS  Google Scholar 

  78. • Penko D, Rojas-Canales D, Mohanasundaram D, Peiris HS, Sun WY, Drogemuller CJ, et al. Endothelial progenitor cells enhance islet engraftment, influence beta cell function and modulate islet connexin 36 expression. Cell Transplant. 2015;24:37–48. https://doi.org/10.3727/096368913X673423 Co-transplantation of endothelial progenitor cells with pancreatic islets enhanced islet engraftment by improving the cure rate and the glucose control.

    Article  PubMed  Google Scholar 

  79. Golshayan D, Pascual M. Tolerance-inducing immunosuppressive strategies in clinical transplantation. Drugs. 2008;68:2113–30. https://doi.org/10.1016/S0140-6736(98)07493-5.

    Article  CAS  PubMed  Google Scholar 

  80. Xing Y, Hogquist KA. T-cell tolerance: central and peripheral. Cold Spring Harb Perspect Biol 2012;1–16.

  81. Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol. 2010;10:490–500. https://doi.org/10.1038/nri2785.

    Article  CAS  PubMed  Google Scholar 

  82. Tang Q, Bluestone JA. Regulatory T-cell therapy in transplantation: moving to the clinic. Cold Spring Harb Perspect Med. 2013;3:1–15. https://doi.org/10.1101/cshperspect.a015552.

    Article  CAS  Google Scholar 

  83. Vignali DAA, Collison LW, Workman CJ. How regulatory T cells work. Nat Rev Immunol. 2008;8:523–32. https://doi.org/10.1038/nri2343.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Cunningham EC, Sharland AF, Alex Bishop G. Liver transplant tolerance and its application to the clinic: can we exploit the high dose effect? Clin Dev Immunol. 2013;2013:1–9. https://doi.org/10.1155/2013/419692.

    Article  CAS  Google Scholar 

  85. Cippà PE, Fehr T. Spontaneous tolerance in kidney transplantation - an instructive, but very rare paradigm. Transpl Int. 2011;24:534–5. https://doi.org/10.1111/j.1432-2277.2011.01260.x.

    Article  PubMed  Google Scholar 

  86. Orlando G, Hematti P, Stratta R, Burke G, Di Cocco P, Pisani F, et al. Clinical operational tolerance after renal transplantation. Ann Surg. 2010;252:915–28. https://doi.org/10.1038/nbt.3121.ChIP-nexus.

    Article  PubMed  Google Scholar 

  87. Li W, Kuhr CS, Zheng XX, Carper K, Thomson AW, Reyes JD, et al. New insights into mechanisms of spontaneous liver transplant tolerance: the role of Foxp3-expressing CD25+CD4+ regulatory T cells. Am J Transplant. 2008;8:1639–51. https://doi.org/10.1111/j.1600-6143.2008.02300.x.

    Article  CAS  PubMed  Google Scholar 

  88. Heidt S, Wood KJ. Biomarkers of operational tolerance in solid organ transplantation. Expert Opin Med Diagn. 2012;6:281–93. https://doi.org/10.1517/17530059.2012.680019.BIOMARKERS.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Niemann N, Sawitzki B. Treg therapy in transplantation: how and when will we do it ? Curr Transplant Reports. 2015;2:233–41. https://doi.org/10.1007/s40472-015-0066-5.

    Article  Google Scholar 

  90. Riley JL, June CH, Blazar BR. Human T regulatory cells as therapeutic agents: take a billion or so of these and call me in the morning. Immunity. 2010;30:656–65. https://doi.org/10.1016/j.immuni.2009.04.006.Human.

    Article  Google Scholar 

  91. McMurchy AN, Bushell A, Levings MK, Wood KJ. Moving to tolerance: clinical application of T regulatory cells. Semin Immunol. 2011;23:304–13. https://doi.org/10.1016/j.smim.2011.04.001.

    Article  CAS  PubMed  Google Scholar 

  92. Xiao F, Ma L, Zhao M, Huang G, Mirenda V, Dorling A, et al. Ex vivo expanded human regulatory T cells delay islet allograft rejection via inhibiting islet-derived monocyte chemoattractant protein-1 production in CD34+ stem cells-reconstituted NOD-scid IL2rgnull mice. PLoS One. 2014;9:1–12. https://doi.org/10.1371/journal.pone.0090387.

    Article  CAS  Google Scholar 

  93. Wu DC, Hester J, Nadig SN, Zhang W, Trzonkowski P, Gray D, et al. Ex vivo expanded human regulatory T cells can prolong survival of a human islet allograft in a humanized mouse model. Transplantation. 2013;96:707–16. https://doi.org/10.1097/TP.0b013e31829fa271.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. • Chandran S, Tang Q, Sarwal M, Polyclonal Regulatory T. Cell therapy for control of inflammation in kidney transplants. Am J Transplant. 2017;17:2945–54. https://doi.org/10.1111/ajt.14415 Infusion of ex vivo expanded regulatory T cells was well tolerated with no adverse effects in kidney transplant recipients.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. • Todo S, Yamashita K, Goto R, Zaitsu M, Nagatsu A, Oura T, et al. A pilot study of operational tolerance with a regulatory T-cell-based cell therapy in living donor liver transplantation. Hepatology. 2016;64:632–43. https://doi.org/10.1002/hep.28459 Infusion of ex vivo expanded regulatory T cells was well tolerated with no adverse effects in liver transplant recipients. Seven patients have been maintaining normal graft function without the use of immunosuppressive drugs for more than 16 months.

    Article  CAS  PubMed  Google Scholar 

  96. Ackermann AM, Gannon M. Molecular regulation of pancreatic beta-cell mass development, maintenance, and expansion. J Mol Endocrinol. 2007;38:193–206. https://doi.org/10.1677/JME-06-0053.

    Article  CAS  PubMed  Google Scholar 

  97. Hughes A, Rojas-canales D, Drogemuller C. IGF2: an endocrine hormone to improve islet transplant survival. J Endocrinol. 2014;221:R41–8. https://doi.org/10.1530/JOE-13-0557.

    Article  CAS  PubMed  Google Scholar 

  98. Reik W, Sun F-L, Dean WL, Kelsey G, Allen ND. Transactivation of Igf2 in a mouse model of Beckwith-Wiedemann syndrome. Nature. 1997;389:809–15. https://doi.org/10.1038/39797.

    Article  CAS  PubMed  Google Scholar 

  99. Petrik J, Arany E, McDonald TJ, Hill DJ. Apoptosis in the pancreatic islet cells of the neonatal rat is associated with a reduced expression of insulin-like growth factor II that may act as a survival factor. Endocrinology. 1998;139:2994–3004. https://doi.org/10.1210/endo.139.6.6042.

    Article  CAS  PubMed  Google Scholar 

  100. Hill DJ, Strutt B, Arany E, Zaina S, Coukell S, Graham CF. Increased and persistent circulating insulin-like growth factor II in neonatal transgenic mice suppresses developmental apoptosis in the pancreatic islets. Endocrinology. 2000;141:1151–7. https://doi.org/10.1210/endo.141.3.7354.

    Article  CAS  PubMed  Google Scholar 

  101. Hogg J, Han VK, Clemmons DR, Hill DJ. Interactions of nutrients, insulin-like growth factors (IGFs) and IGF-binding proteins in the regulation of DNA synthesis by isolated fetal rat islets of Langerhans. J Endocrinol. 1993;138:401–12. https://doi.org/10.1677/joe.0.1380401.

    Article  CAS  PubMed  Google Scholar 

  102. Robitaille R, Dusseault J, Henley N, Rosenberg L, Hallé J-P. Insulin-like growth factor II allows prolonged blood glucose normalization with a reduced islet cell mass transplantation. Endocrinology. 2003;144:3037–45. https://doi.org/10.1210/en.2002-0185.

    Article  CAS  PubMed  Google Scholar 

  103. Petrik J, Reusens B, Arany E, Remacle C, Coelho C, Hoet JJ, et al. A low protein diet alters the balance of islet cell replication and apoptosis in the fetal and neonatal rat and is associated with a reduced pancreatic expression of insulin-like growth factor-II. Endocrinology. 1999;140:4861–73. https://doi.org/10.1210/endo.140.10.7042.

    Article  CAS  PubMed  Google Scholar 

  104. Hughes A, Mohanasundaram D, Kireta S, Jessup CF, Drogemuller CJ, Coates PTH. Insulin-like growth factor-II (IGF-II) prevents proinflammatory cytokine-induced apoptosis and significantly improves islet survival after transplantation. Transp J. 2013;95:671–8. https://doi.org/10.1097/TP.0b013e31827fa453.

    Article  CAS  Google Scholar 

  105. Shibuya M. Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling in angiogenesis: a crucial target for anti- and pro-angiogenic therapies. Genes and Cancer. 2011;2:1097–105. https://doi.org/10.1177/1947601911423031.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Lammert E, Gu G, Mclaughlin M, Brown D, Brekken R, Murtaugh LC, et al. Role of VEGF-A in vascularization of pancreatic islets. Curr Biol. 2003;13:1070–4. https://doi.org/10.1016/S0960-9822(03)00378-6.

    Article  CAS  PubMed  Google Scholar 

  107. Witkowski P, Sondermeijer H, Hardy MA, Woodland DC, Lee K, Bhagat G, et al. Islet grafting and imaging in a bioengineered intramuscular space. Transplantation. 2009;88:1065–74. https://doi.org/10.1097/TP.0b013e3181ba2e87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. • Marchioli G, Di Luca A, De Koning E, Engelse M, Van Blitterswijk CA, Karperien M, et al. Hybrid polycaprolactone/alginate scaffolds functionalized with VEGF to promote de novo vessel formation for the transplantation of islets of Langerhans. Adv Healthc Mater. 2016;5:1606–16. https://doi.org/10.1002/adhm.201600058 Polycaprolactone scaffold functionalized with VEGF promoted vascularization and improved function of encapsulated islets embedded within the polycaprolactone scaffold.

    Article  CAS  PubMed  Google Scholar 

  109. Boyman O, Sprent J. The role of interleukin - 2 during homeostasis and activation of the immune system. Nat Rev Immunol. 2012;12:180–90. https://doi.org/10.1038/nri3156.

    Article  CAS  PubMed  Google Scholar 

  110. Sakaguchi S, Takahashi T, Nishizuka Y. Study on cellular events in post-thymectomy autoimmune oophoritis in mice. J Exp Med. 1982;156:1577–86.

    Article  CAS  PubMed  Google Scholar 

  111. Chinen T, Kannan AK, Levine AG, Fan X, Klein U, Zheng Y, et al. An essential role for IL-2 receptor in regulatory T cell function Takatoshi. Nat Immunol. 2016;17:1322–33. https://doi.org/10.1038/ni.3540.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Létourneau S, Krieg C, Pantaleo G, Boyman O. IL-2- and CD25-dependent immunoregulatory mechanisms in the homeostasis of T-cell subsets. J Allergy Clin Immunol. 2009;123:758–62. https://doi.org/10.1016/j.jaci.2009.02.011.

    Article  CAS  PubMed  Google Scholar 

  113. Sadlon TJ, Wilkinson BG, Pederson S, Brown CY, Bresatz S, Gargett T, et al. Genome-wide identification of human FOXP3 target genes in natural regulatory T cells. J Immunol. 2010;185:1071–81. https://doi.org/10.4049/jimmunol.1000082.

    Article  CAS  PubMed  Google Scholar 

  114. Beyer M, Thabet Y, Müller R, Sadlon T, Classen S, Lahl K, et al. Repression of the genome organizer SATB1 in regulatory T cells is required for suppressive function and inhibition of effector differentiation. Nat Immunol. 2011;12:898–907. https://doi.org/10.1038/ni.2084.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Whitehouse G, Gray E, Mastoridis S, Merritt E, Kodela E, Yang JHM. IL-2 therapy restores regulatory T-cell dysfunction induced by calcineurin inhibitors. Proc Natl Acad Sci USA. 2017;114:7083–8. https://doi.org/10.1073/pnas.1620835114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Shevach EM. Application of IL-2 therapy to target T regulatory cell function. Trends Immunol. 2013;33:626–32. https://doi.org/10.1016/j.it.2012.07.007.

    Article  CAS  Google Scholar 

  117. Zorn E, Nelson EA, Mohseni M, Porcheray F, Kim H, Litsa D, et al. IL-2 regulates FOXP3 expression in human CD4+ CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood. 2006;108:1571–80. https://doi.org/10.1182/blood-2006-02-004747.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Koreth J, Matsuoka K, Kim HT, McDonough SM, Bindra B, Alyea EP, et al. Interleukin-2 and regulatory T cells in graft-versus-host disease. N Engl J Med. 2011;365:2055–66. https://doi.org/10.1056/NEJMoa1108188.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Matsuoka K, Koreth J, Kim HT, Bascug OG, Kawano Y, Murase K, et al. Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease. Sci Transl Med. 2013;5:179ra43. https://doi.org/10.1126/scitranslmed.3005265.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Koreth J, Kim HT, Jones KT, Lange PB, Reynolds CG, Chammas MJ, et al. Efficacy , durability , and response predictors of low-dose interleukin-2 therapy for chronic graft-versus-host disease. Blood. 2016;128:130–8. https://doi.org/10.1182/blood-2016-02-702852.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to P. Toby Coates.

Ethics declarations

Conflict of Interest

The authors declare that they have no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Immunology, Transplantation, and Regenerative Medicine

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, J., Kang, K., Drogemuller, C.J. et al. Bioprinting an Artificial Pancreas for Type 1 Diabetes. Curr Diab Rep 19, 53 (2019). https://doi.org/10.1007/s11892-019-1166-x

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11892-019-1166-x

Keywords

Navigation