Micro-fabricated scaffolds lead to efficient remission of diabetes in mice

Mijke Buitinga, Frank Assen, Maaike Hanegraaf, Paul Wieringa, Janneke Hilderink, Lorenzo Moroni, Roman Truckenmüller, Clemens van Blitterswijk, Gert-Willem Roemer, Francoise Carlotti, Eelco de Koning, Marcel Karperien, Aart van Apeldoorn*

*Corresponding author for this work

Research output: Contribution to journalArticleAcademicpeer-review

161 Downloads (Pure)

Abstract

Despite the clinical success of intrahepatic islet transplantation in treating type 1 diabetes, factors specific to this transplantation site hinder long-term insulin independence. The adoption of alternative, extravascular sites likely improve islet survival and function, but few locations are able to sufficiently confine islets in order to facilitate engraftment. This work describes a porous microwell scaffold with a well-defined pore size and spacing designed to guarantee islet retention at an extrahepatic transplantation site and facilitate islet revascularization. Three techniques to introduce pores were characterized: particulate leaching; solvent casting on pillared wafers; and laser drilling. Our criteria of a maximum pore diameter of 40 mm were best achieved via laser drilling. Transplantation studies in the epididymal fat of diabetic mice elucidated the potential of this porous scaffold platform to restore blood glucose levels and facilitate islet engraftment. Six out of eight mice reverted to stable normoglycemia with a mean time to remission of 6.2 +/- 3.2 days, which was comparable to that of the gold standard of renal subcapsular islet grafts. In contrast, when islets were transplanted in the epididymal fat pad without a microwell scaffold, only two out of seven mice reverted to stable normoglycemia. Detailed histological evaluation four weeks after transplantation found a comparable vascular density in scaffold-seeded islets, renal subcapsular islets and native pancreatic islets. However, the vascularization pattern in scaffold-seeded islets was more inhomogeneous compared to native pancreatic islets with a higher vascular density in the outer shell of the islets compared to the inner core. We also observed a corresponding decrease in the beta-cell density in the islet core. Despite this, our data indicated that islets transplanted in the microwell scaffold platform were able to maintain a viable beta-cell population and restore glycemic control. Furthermore, we demonstrated that the microwell scaffold platform facilitated detailed analysis at a subcellular level to correlate design parameters with functional physiological observations. (C) 2017 Elsevier Ltd. All rights reserved.

Original languageEnglish
Pages (from-to)10-22
Number of pages13
JournalBiomaterials
Volume135
DOIs
Publication statusPublished - Aug 2017

Keywords

  • Diabetes
  • Islet
  • Transplantation
  • Microwell scaffolds
  • EXTRAHEPATIC ISLET TRANSPLANTATION
  • PANCREATIC-ISLETS
  • BETA-CELLS
  • POLYMER SCAFFOLDS
  • ENGRAFTMENT
  • TISSUE
  • HYPOXIA
  • SITE
  • VASCULARIZATION
  • BIOMATERIALS

Cite this